WO2014197748A2 - Édition et régulation géniques à guidage arn - Google Patents

Édition et régulation géniques à guidage arn Download PDF

Info

Publication number
WO2014197748A2
WO2014197748A2 PCT/US2014/041190 US2014041190W WO2014197748A2 WO 2014197748 A2 WO2014197748 A2 WO 2014197748A2 US 2014041190 W US2014041190 W US 2014041190W WO 2014197748 A2 WO2014197748 A2 WO 2014197748A2
Authority
WO
WIPO (PCT)
Prior art keywords
gene
target
cell
cas9
different
Prior art date
Application number
PCT/US2014/041190
Other languages
English (en)
Other versions
WO2014197748A3 (fr
Inventor
Charles A. GERSBACH
Isaac B. HILTON
Pablo Perez-Pinera
Ami M. KABADI
Pratiksha I. THAKORE
David G. OUSTEROUT
Joshua B. BLACK
Original Assignee
Duke University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CA2914519A priority Critical patent/CA2914519A1/fr
Priority to CN202111496852.0A priority patent/CN114230675A/zh
Priority to EP18172956.7A priority patent/EP3417880A1/fr
Priority to KR1020237022139A priority patent/KR20230107387A/ko
Priority to US14/895,316 priority patent/US10704060B2/en
Priority to EP19168481.0A priority patent/EP3539573B1/fr
Priority to KR1020167000166A priority patent/KR102307280B1/ko
Priority to JP2016518017A priority patent/JP7085716B2/ja
Priority to CN201480044748.XA priority patent/CN105658805B/zh
Priority to KR1020217017616A priority patent/KR102551324B1/ko
Priority to AU2014274840A priority patent/AU2014274840B2/en
Priority to KR1020237031879A priority patent/KR20230136697A/ko
Application filed by Duke University filed Critical Duke University
Priority to EP14806852.1A priority patent/EP3004370A4/fr
Publication of WO2014197748A2 publication Critical patent/WO2014197748A2/fr
Publication of WO2014197748A3 publication Critical patent/WO2014197748A3/fr
Priority to US15/991,333 priority patent/US10745714B2/en
Priority to US16/858,689 priority patent/US20210002665A1/en
Priority to AU2020203924A priority patent/AU2020203924A1/en
Priority to US16/927,679 priority patent/US20210032654A1/en
Priority to JP2022052186A priority patent/JP2022101562A/ja
Priority to AU2022250572A priority patent/AU2022250572A1/en
Priority to AU2022250450A priority patent/AU2022250450A1/en
Priority to JP2023131956A priority patent/JP2023164823A/ja

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4707Muscular dystrophy
    • C07K14/4708Duchenne dystrophy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1003Transferases (2.) transferring one-carbon groups (2.1)
    • C12N9/1007Methyltransferases (general) (2.1.1.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/71Fusion polypeptide containing domain for protein-protein interaction containing domain for transcriptional activaation, e.g. VP16
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/40Systems of functionally co-operating vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)

Definitions

  • the present disclosure relates to the field of gene expression alteration, genome engineering and genomic alteration of genes using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) 9-based systems and viral delivery systems.
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • Cas CRISPR-associated 9-based systems and viral delivery systems.
  • the present disclosure also relates to the field of genome engineering and genomic alteration of genes in muscle, such as skeletal muscle and cardiac muscle.
  • Synthetic transcription factors have been engineered to control gene expression for many different medical and scientific applications in mammalian systems, including stimulating tissue regeneration, drug screening, compensating for genetic defects, activating silenced tumor suppressors, controlling stem cell differentiation, performing genetic screens, and creating synthetic gene circuits.
  • These transcription factors can target promoters or enhancers of endogenous genes, or be purposefully designed to recognize sequences orthogonal to mammalian genomes for transgene regulation.
  • the most common strategies for engineering novel transcription factors targeted to user-defined sequences have been based on the programmable DNA-binding domains of zinc finger proteins and transcription-activator like effectors (TALEs). Both of these approaches involve applying the principles of protein-DNA interactions of these domains to engineer new proteins with unique DNA-binding specificity. Although these methods have been widely successful for many applications, the protein engineering necessary for manipulating protein-DNA interactions can be laborious and require specialized expertise.
  • these new proteins are not always effective. The reasons for this are not yet known but may be related to the effects of epigenetic modifications and chromatin state on protein binding to the genomic target site. In addition, there are challenges in ensuring that these new proteins, as well as other components, are delivered to each cell.
  • Existing methods for delivering these new proteins and their multiple components include delivery to cells on separate plasmids or vectors which leads to highly variable expression levels in each cell due to differences in copy number. Additionally, gene activation following transfection is transient due to dilution of plasmid DNA, and temporary gene expression may not be sufficient for inducing therapeutic effects. Furthermore, this approach is not amenable to cell types that are not easily transfected. Thus another limitation of these new proteins is the potency of transcriptional activation.
  • Site-specific nucleases can be used to introduce site-specific double strand breaks at targeted genomic loci. This DNA cleavage stimulates the natural DNA-repair machinery, leading to one of two possible repair pathways. In the absence of a donor template, the break will be repaired by non-homologous end joining (NHEJ), an error-prone repair pathway that leads to small insertions or deletions of DNA. This method can be used to intentionally disrupt, delete, or alter the reading frame of targeted gene sequences. However, if a donor template is provided along with the nucleases, then the cellular machinery will repair the break by homologous recombination, which is enhanced several orders of magnitude in the presence of DNA cleavage.
  • NHEJ non-homologous end joining
  • Engineered nucleases have been used for gene editing in a variety of human stem cells and cell lines, and for gene editing in the mouse liver.
  • the major hurdle for implementation of these technologies is delivery to particular tissues in vivo in a way that is effective, efficient, and facilitates successful genome modification.
  • Hereditary genetic diseases have devastating effects on children in the United States. These diseases currently have no cure and can only be managed by attempts to alleviate the symptoms. For decades, the field of gene therapy has promised a cure to these diseases.
  • DMD Duchenne Muscular Dystrophy
  • DMD Duchenne Muscular Dystrophy
  • Dystrophin is a key component of a protein complex that is responsible for regulating muscle cell integrity and function.
  • DMD patients typically lose the ability to physically support themselves during childhood, become progressively weaker during the teenage years, and die in their twenties.
  • Current experimental gene therapy strategies for DMD require repeated administration of transient gene delivery vehicles or rely on permanent integration of foreign genetic material into the genomic DNA. Both of these methods have serious safety concerns. Furthermore, these strategies have been limited by an inability to deliver the large and complex dystrophin gene sequence.
  • the present invention is directed to a fusion protein comprising two heterologous polypeptide domains.
  • the first polypeptide domain comprises a Clustered Regularly Interspaced Short Palindromic Repeats associated (Cas) protein and the second polypeptide domain has an activity selected from the group consisting of transcription activation activity, transcription repression activity, transcription release factor activity, histone modification activity, nuclease activity, nucleic acid association activity, methylase activity, and demethylase activity.
  • the Cas protein may comprise Cas9.
  • the Cas9 may comprise at least one amino acid mutation which knocks out nuclease activity of Cas9.
  • the at least one amino acid mutation may be at least one of D10A and H840A.
  • the Cas protein may comprise iCas9 (amino acids 36-1403 of SEQ ID NO: 1).
  • the second polypeptide domain may have transcription activation activity.
  • the second polypeptide domain may comprise at least one VP 16 transcription activation domain repeat.
  • the second polypeptide domain may comprise a VP 16 tetramer ("VP64") or a p65 activation domain.
  • the fusion protein may further comprise a linker connecting the first polypeptide domain to the second polypeptide domain.
  • the fusion protein may comprise iCas9-VP64.
  • the present invention is directed to a DNA targeting system comprising said fusion protein and at least one guide RNA (gRNA).
  • the at least one gRNA may comprise a 12-22 base pair complementary polynucleotide sequence of the target DNA sequence followed by a protospacer-adjacent motif.
  • the at least one gRNA may target a promoter region of a gene, an enhancer region of a gene, or a transcribed region of a gene.
  • the at least one gRNA may target an intron of a gene.
  • the at least one gRNA may target an exon of a gene.
  • the at least one gRNA may target a the promoter region of a gene selected from the group consisting of ASCL1, BRN2, MYTIL, NANOG, VEGFA, TERT, ILIB, IL1R2, ILIRN, HBGl, HBG2, and MYODI.
  • the at least one gRNA may comprise at least one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, and 585-625.
  • the present invention is directed to a DNA targeting system that binds to a dystrophin gene comprising Cas9 and at least one guide RNA (gRNA).
  • the at least one gRNA may target an intron of the dystrophin gene.
  • the at least one gRNA may target an exon of the dystrophin gene.
  • the at least one guide RNA may comprise at least one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, and 585-625.
  • the DNA targeting system may comprise between one and ten different gRNAs.
  • the present invention is directed to an isolated polynucleotide encoding said fusion protein or said DNA targeting system.
  • the present invention is directed to a vector comprising said isolated polynucleotide.
  • the present invention is directed to a cell comprising said isolated polynucleotide or said vector.
  • the present invention is directed to a method of modulating mammalian gene expression in a cell.
  • the method comprises contacting the cell with said fusion protein, said DNA targeting system, said isolated polynucleotide, or said vector.
  • the gene expression may be induced.
  • the present invention is directed to a method of transdifferentiating or inducing differentiation of a cell.
  • the method comprises contacting the cell with said fusion protein, said DNA targeting system, said isolated polynucleotide, or said vector.
  • the cell may be a fibroblast cell or an induced pluripotent stem cells.
  • the fibroblast cell may be trandifferentiated into a neuronal cell or a myogenic cell.
  • the DNA targeting system may be contacted with the cell and at least one gRNA targets a promoter region of at least one gene selected from the group consisting of ASCL1, BRN2, MYOD1, and MYT1L.
  • the DNA targeting system may comprise at least one gR A that targets the promoter region of the ASCL1 gene and at least one gR A that targets the promoter region of the BRN2 gene.
  • the DNA targeting system may comprise between one and twenty different gRNAs.
  • the DNA targeting system may comprise 8 or 16 different gRNAs.
  • the DNA targeting system may comprise dCas9-VP64.
  • the DNA targeting system may be delivered to the cell virally or non-virally.
  • the present invention is directed to a method of correcting a mutant gene in a cell.
  • the method comprises administering to a cell containing said DNA targeting system, said isolated polynucleotide, or said vector.
  • the correction of the mutant gene may comprise homology-directed repair.
  • the method may further comprise administering to the cell a donor DNA.
  • the mutant gene may comprise a frameshift mutation which causes a premature stop codon and a truncated gene product.
  • the correction of the mutant gene may comprise nuclease mediated non-homologous end joining.
  • the correction of the mutant gene may comprise a deletion of a premature stop codon, a disruption of a splice acceptor site, a deletion of one or more exons, or disruption of a splice donor sequence.
  • the deletion of one or more exons may result in the correction of the reading frame.
  • the present invention is directed to a method of treating a subject in need thereof having a mutant dystrophin gene.
  • the method comprises administering to the subject said DNA targeting system, said isolated polynucleotide, or said vector.
  • the subject may be suffering from Duchenne muscular dystrophy.
  • the present invention is directed to a method of correcting a mutant dystrophin gene in a cell.
  • the method comprises administering to a cell containing a mutant dystrophin gene said DNA targeting system, said isolated polynucleotide, said vector, or said cell.
  • the mutant dystrophin gene may comprise a premature stop codon, disrupted reading frame via gene deletion, an aberrant splice acceptor site, or an aberrant splice donor site, and wherein the target region is upstream or downstream of the premature stop codon, disrupted reading frame, aberrant splice acceptor site, or the aberrant splice donor site.
  • the correction of the mutant dystrophin gene may comprise homology-directed repair.
  • the method may further comprise administering to the cell a donor DNA.
  • the mutant dystrophin gene may comprise a frameshift mutation which causes a premature stop codon and a truncated gene product.
  • the correction of the mutant dystrophin gene may comprise nuclease mediated non-homologous end joining.
  • the correction of the mutant dystrophin gene may comprise a deletion of a premature stop codon, correction of a disrupted reading frame, or modulation of splicing by disruption of a splice acceptor site or disruption of a splice donor sequence.
  • the correction of the mutant dystrophin gene may comprise a deletion of exons 45-55 or exon 51.
  • the present invention is directed to a kit comprising said fusion protein, said DNA targeting system, said isolated polynucleotide, said vector, or said cell.
  • the present invention is directed to a method of modulating mammalian gene expression in a cell.
  • the method comprises contacting the cell with a polynucleotide encoding a DNA targeting system.
  • the DNA targeting system comprises said fusion protein and at least one guide RNA (gRNA).
  • the DNA targeting system may comprise between one and ten different gRNAs.
  • the different gRNAs may bind to different target regions within the target gene.
  • the target regions may be separated by at least one nucleotide.
  • the target regions may be separated by about 15 to about 700 base pairs.
  • Each of the different gRNAs may bind to at least one different target genes.
  • the different target genes may be located on same chromosome.
  • the different target genes may be located on different chromosomes.
  • the at least one target region may be within a non-open chromatin region, an open chromatin region, a promoter region of the target gene, an enhancer region of the target gene, a transcribed region of the target gene, or a region upstream of a transcription start site of the target gene.
  • the at least one target region may be located between about 1 to about 1000 base pairs upstream of a transcription start site of a target gene.
  • the at least one target region may be located between about 1 to about 600 base pairs upstream of a transcription start site of a target gene.
  • the gene expression may be induced.
  • the DNA targeting system may comprise two different gRNAs, three different gRNAs, four different gRNAs, five different gRNAs, six different gRNAs, seven different gRNAs, eight different gRNAs, nine different gRNAs, or ten different gRNAs.
  • the at least one guide RNA may target a promoter region of a gene selected from the group consisting of ASCL1, BRN2, MYT1L, NANOG, VEGFA, TERT, IL1B, IL1R2, IL1RN, HBG1, HBG2, and MYOD1.
  • the at least one guide RNA may comprise at least one of SEQ ID NOs: 5-40, 65-144, 492-515, 540- 563, and 585-625.
  • the at least one target region may be within an intron or an exon of a target gene.
  • the present invention is directed to a composition for inducing mammalian gene expression in a cell.
  • the composition comprises said fusion protein and at least one guide RNA (gRNA).
  • the present invention is directed to a composition for inducing mammalian gene expression in a cell.
  • the composition comprises an isolated polynucleotide sequence encoding said fusion protein and at least one guide RNA (gRNA).
  • the at least one guide RNA may target a promoter region of a gene selected from the group consisting of ASCL1, BRN2, MYT1L, NANOG, VEGFA, TERT, IL1B, IL1R2, IL1RN, HBG1, HBG2, and MYOD1.
  • the at least one guide RNA may comprise at least one of SEQ ID NOs5-40, 65-144, 492-515, 540-563, and 585- 625.
  • the present invention is directed to a cell comprising said composition for inducing mammalian gene expression in a cell.
  • the present invention is directed to a kit comprising said composition for inducing mammalian gene expression in a cell or said cell comprising said composition for inducing mammalian gene expression in a cell.
  • the present invention is directed to a kit for inducing mammalian gene expression in a cell.
  • the kit comprises said composition for inducing mammalian gene expression in a cell or said cell comprising said composition for inducing mammalian gene expression in a cell.
  • the present invention is directed to a composition for genome editing in a muscle of a subject.
  • the composition comprises a modified adeno-associated virus (AAV) vector and a nucleotide sequence encoding a site-specific nuclease.
  • the muscle is skeletal muscle or cardiac muscle.
  • the modified AAV vector may have enhanced cardiac and skeletal muscle tissue tropism.
  • the site-specific nuclease may comprise a zinc finger nuclease, a TAL effector nuclease, or a CRISPR/Cas9 system.
  • the site-specific nuclease may bind a gene or locus in the cell of the muscle.
  • the gene or locus may be dystrophin gene.
  • the composition may further comprise a donor DNA or transgene.
  • the present invention is directed to a kit comprising said composition for genome editing in a muscle of a subject.
  • the present invention is directed to a method of genome editing in a muscle of a subject.
  • the method comprises administering to the muscle said composition for genome editing in a muscle of a subject, wherein the muscle is skeletal muscle or cardiac muscle.
  • the genome editing may comprise correcting a mutant gene or inserting a transgene. Correcting a mutant gene may comprise deleting, rearranging, or replacing the mutant gene. Correcting the mutant gene may comprise nuclease-mediated non-homologous end joining or homology-directed repair.
  • the present invention is directed to a method of treating a subject.
  • the method comprises administering said composition for genome editing in a muscle of a subject to a muscle of the subject, wherein the muscle is skeletal muscle or cardiac muscle.
  • the subject may be suffering from a skeletal muscle condition or a genetic disease.
  • the subject may be suffering from Duchenne muscular dystrophy.
  • the present invention is directed to a method of correcting a mutant gene in a subject, the method comprises administering said composition for genome editing in a muscle of a subject.
  • the muscle is skeletal muscle or cardiac muscle.
  • the composition may be injected into the skeletal muscle of the subject.
  • the composition may be injected systemically to the subject.
  • the skeletal muscle may be tibialis anterior muscle.
  • the present invention is directed to a modified lentiviral vector for genome editing in a subject comprising a first polynucleotide sequence encoding said fusion protein and a second polynucleotide sequence encoding at least one sgRNA.
  • the first polynucleotide sequence may be operably linked to a first promoter.
  • the first promoter may be a constitutive promoter, an inducible promoter, a repressible promoter, or a regulatable promoter.
  • the second promoter may be a constitutive promoter, an inducible promoter, a repressible promoter, or a regulatable promoter.
  • polynucleotide sequence may encode between one and ten different sgRNAs.
  • the second polynucleotide sequence may encode two different sgRNAs, three different sgRNAs, four different sgRNAs, five different sgRNAs, six different sgRNAs, seven different sgRNAs, eight different sgRNAs, nine different sgRNAs, or ten different sgRNAs.
  • Each of the polynucleotide sequences encoding the different sgRNAs may be operably linked to a promoter.
  • Each of the promoters operably linked to the different sgRNAs may be the same promoter.
  • Each of the promoters operably linked to the different sgRNAs may be different promoters.
  • the promoter may be a constitutive promoter, an inducible promoter, a repressible promoter, or a regulatable promoter.
  • the sgRNA may bind to a target gene. Each of the sgRNA may bind to a different target region within one target loci. Each of the sgRNA may bind to a different target region within different gene loci.
  • the fusion protein may comprise Cas9 protein or iCas9-VP64 protein.
  • the fusion protein may comprise a VP64 domain, a p300 domain, or a KRAB domain.
  • the two or more endogenous genes may be transcriptionally activated. The two or more endogenous genes may be repressed.
  • the present invention is directed to a method of activating an endogenous gene in a cell.
  • the method comprises contacting a cell with said modified lentiviral vector.
  • the endogenous gene may be transiently activated.
  • the endogenous gene may be stably activated.
  • the endogenous gene may be transiently repressed.
  • the endogenous gene may be stably repressed.
  • the fusion protein may be expressed at similar levels to the sgRNAs.
  • the fusion protein may be expressed at different levels to the sgRNAs.
  • the cell may be a primary human cell.
  • the present invention is directed to a method of multiplex gene editing in a cell.
  • the method comprises contacting a cell with said modified lentiviral vector.
  • the multiplex gene editing may comprise correcting at least one mutant gene or inserting a transgene.
  • Correcting a mutant gene may comprise deleting, rearranging, or replacing the at least one mutant gene.
  • Correcting the at least one mutant gene may comprise nuclease-mediated non-homologous end joining or homology-directed repair.
  • the multiplex gene editing may comprise deleting at least one gene, wherein the gene is an endogenous normal gene or a mutant gene.
  • the multiplex gene editing may comprise deleting at least two genes.
  • the multiplex gene editing may comprise deleting between two and ten genes.
  • the present invention is directed to a method of modulating gene expression of at least one target gene in a cell.
  • the method comprises contacting a cell with said modified lentiviral vector.
  • the gene expression of at least two genes may be modulated.
  • the gene expression of between two genes and ten genes may be modulated.
  • the gene expression of the at least one target gene may be modulated when gene expression levels of the at least one target gene are increased or decreased compared to normal gene expression levels for the at least one target gene.
  • Fig. 1 shows RNA-guided activation of the human IL1RN gene by iCas9-VP64.
  • a,b An RNA-guided transcriptional activator was created by fusing the inactivated Cas9 (iCas9, D10A/H840A) to the VP64 transactivation domain.
  • iCas9-VP64 recognizes genomic target sites through the hybridization of a guide RNA (gRNA) to a 20 bp target sequence
  • gRNA guide RNA
  • Expression plasmids for four gR As or crR A/tracrRNAs targeted to sequences in the IL1RN promoter were co-transfected with the iCas9-VP64 expression plasmid into HEK293T cells. Activation of IL1RN expression was assessed by qRT-PCR.
  • the four gRNA expression plasmids were co- transfected with iCas9-VP64 individually or in combination. Robust gene activation was observed by qRT-PCR only in response to the combination of gRNAs.
  • FIG. 2 shows RNA-guided activation of human genes relevant to cell and gene therapy, genetic reprogramming, and regenerative medicine.
  • HEK293T cells were transfected with the iCas9-VP64 expression plasmid and four gRNAs individually or in combination.
  • FIG. 3 shows expression of iCas9-VP64. Expression of iCas9-VP64 in transfected HEK293 cells was confirmed by western blot for the N-terminal Flag epitope tag. The wt Cas9 expression plasmid does not contain the epitope tag.
  • Fig. 4 shows positions of gRNA target sites and DNAse hypersensitivity of human target genes.
  • the four gRNA target sites for each locus are designated as custom tracks above each gene and DNase-seq data indicating DNAse-hypersensitive open chromatin regions is shown below each gene.
  • DNase-seq was performed in HEK293T cells to identify DNase hypersensitive regions, as previously described (Song et al, Cold Spring Harbor protocols 2010, pdb prot5384 (2010); Song et al. Genome Res 21, 1757-1767 (2011)). The results show that open chromatin was not a requirement for gene activation by combinations of gRNAs with iCas9-VP64.
  • Fig. 4 shows positions of gRNA target sites and DNAse hypersensitivity of human target genes.
  • the four gRNA target sites for each locus are designated as custom tracks above each gene and DNase-seq data indicating DNAse-hypersensitive open chromatin regions is shown below each gene.
  • FIG. 5 shows the absence of nuclease activity by iCas9-VP64.
  • Wild-type Cas9 or inactivated (D10A, H840A) iCas9-VP64 expression plasmids were co-transfected with expression plasmids for four different guide RNAs targeting the IL1RN promoter.
  • Nuclease activity was determined by the Surveyor assay (Guschin et al, Methods Mol Biol 649, 247-256 (2010)).
  • the lower molecular weight bands indicative of nuclease activity and DNA repair by non-homologous end joining are only present following treatment with wild-type Cas9, supporting abrogation of nuclease activity by iCas9-VP64.
  • Fig. 6 shows RNA-seq for samples treated with gRNAs targeting HBG1 and HBG2.
  • Three of these gRNAs also target HBG2.
  • Fig. 7 shows upregulation of Ascll and ⁇ -globin by iCas9-VP64.
  • HEK293T cells were transfected with iCas9-VP64 and four gRNAs targeting the ASCLI or HBG1 promoter.
  • Levels of corresponding Ascll and ⁇ -globin protein production were assessed by western blot. Low levels of these proteins were detectable in HEK293T cells and increases in expression were detectable following iCas9-VP64 treatment in two independent experiments.
  • FIG. 8 shows activation of downstream targets of Ascll in iCas9-VP64-treated murine embryonic fibroblasts.
  • Mouse embryonic fibroblasts (MEFs) were transfected with a control GFP expression plasmid or the iCas9-VP64 expression plasmid and a combination of four gRNA expression plasmids targeting ASCLI at a ratio of 50:50 or 75:25.
  • the gRNA target sites in the human ASCLI promoter SEQ ID NO: 3 are conserved in the mouse ASCLI promoter (SEQ ID NO: 4).
  • Target sites are indicated by solid lines and the transcribed region is indicated by dashed line
  • ASCLI expression in MEFs increased at two days after iCas9-VP64/gRNA treatment as determined by qRT-PCR.
  • c-h After 10 days in neural induction media, cells were stained for Ascll and Tujl, an early marker of neuronal differentiation (c-d), or for Tuj l and MAP2, a marker of more mature neuronal differentiation (d-f).
  • Some Tuj 1 -positive cells adopted neuronal morphologies (f-g) and a single cell was found to be positive for Tuj 1 and MAP2 (g).
  • Fig. 9 shows (a) the iCas9-VP64 protein sequence (SEQ ID NO: 1) and (b) the sequence of the gRNA expression cassette with U6 promoter (SEQ ID NO: 2).
  • Fig. 10 shows the standard curves for qRT-PCR.
  • the experimental sample with the highest expression level was diluted to create a standard curve that was assayed by qRT-PCR to ensure efficient amplification over an appropriate dynamic range.
  • the efficiencies of all amplification reactions were within 90-115%.
  • Figs. 11(a)- 11(b) show the validation of RNA-guided repair.
  • Fig. 11(a) shows the Surveyor assay results of genomic DNA harvested from HEK 293T cells two days after Cas9 was co-transfected into the cells with empty vector (negative control) or gRNA.
  • Fig. 11(b) shows the location of the gRNA target.
  • Fig. 11(c) shows the expected cleavage sizes for each gRNA.
  • Fig. 12 shows RNA-guided repair in DMD 8036 (del48-50) cells as shown by
  • Fig. 13 shows RNA-guided repair in DMD 8036 (del48-50) cells as shown by PCR across the entire locus.
  • the PCR of a wild-type dystrophin gene generates a fragment of 1447 bp in size, whereas PCR of the mutant gene in the DMD 8036 cell line shows a deletion of approximately 817 bp.
  • the deletion band after introduction of the CRISPR/Cas9-based system was approximately 630 bp.
  • Fig. 14 shows RNA-guided repair in DMD 8036 (del48-50) cells as shown by Western blot with MANDYS8 (anti-dystrophin antibody) and GAPDH antibody (positive control).
  • FIG. 15 shows ChIP sequencing data illustrating the specific binding of iCas9-VP64 targeting the ILIRN promoter.
  • HEK 293T cells were transfected with iCas9-VP64 targeting the ILIRN promoter.
  • Fig. 16 shows CRISPR/Cas9 targeting of the dystrophin gene.
  • sgRNA sequences were designed to bind sequences in the exon 45-55 mutational hotspot region of the dystrophin gene, such that gene editing could restore dystrophin expression from a wide variety of patient- specific mutations. Arrows within introns indicate sgRNA targets designed to delete entire exons from the genome. Arrows within exons indicate sgRNA targets designed to create targeted frameshifts in the dystrophin gene.
  • B Example of frame correction following introduction of small insertions or deletions by NHEJ DNA repair in exon 51 using the CR3 sgRNA.
  • C Schematic of multiplex sgRNA targets designed to delete exon 51 and restore the dystrophin reading frame in a patient mutation with the deletion of exons 48-50.
  • D Schematic of multiplex sgRNA targets designed to delete the entire exon 45-55 region to address a variety of DMD patient mutations.
  • Fig. 17 shows images of TBE-PAGE gels used to quantify Surveyor assay results to measure day 3 gene modification in Table 7. Asterisks mark expected sizes of bands indicative of nuclease activity.
  • Fig. 18 shows images of TBE-PAGE gels used to quantify Surveyor assay results to measure day 10 gene modification in Table 7. Asterisks mark expected sizes of bands indicative of nuclease activity.
  • Fig. 19 shows fluorescence-activated flow sorting to enrich genetically modified DMD myoblasts.
  • a plasmid expressing a human-codon optimized SpCas9 protein linked to a GFP marker using a T2A ribosomal skipping peptide sequence was co-electroporated into human DMD myoblasts with one or two plasmids carrying sgRNA expression cassettes.
  • B The indicated sgRNA expression cassettes were independently co-transfected into HEK293Ts with a separate plasmid expressing SpCas9 with (bottom) or without (top) a GFP marker linked to SpCas9 by a T2A ribosomal skipping peptide sequence.
  • Gene modification frequencies were assessed at 3 days post-transfection by the Surveyor assay.
  • C DMD myoblasts with deletions of exons 48-50 in the dystrophin gene were treated with sgRNAs that correct the dystrophin reading frame in these patient cells. Gene modification was assessed at 20 days post- electroporation in unsorted (bulk) or GFP+ sorted cells.
  • D GFP expression in DMD myoblasts 3 days after electroporation with indicated expression plasmids. Transfection efficiencies and sorted cell populations are indicated by the gated region.
  • Fig. 20 shows targeted frameshifts to restore the dystrophin reading frame using CRISPR/Cas9.
  • A The 5' region of exon 51 was targeted using a sgRNA, CR3, that binds immediately upstream of the first out-of- frame stop codon. PAM: protospacer-adjacent motif.
  • B The exon 51 locus was PCR amplified from HEK293T cells treated with SpCas9 and CR3 expression cassettes. Sequences of individual clones were determined by Sanger sequencing. The top sequence (bolded, exon in red) is the native, unmodified sequence. The number of clones for each sequence is indicated in parentheses.
  • Fig. 21 shows deletion of exon 51 from the human genome using multiplex
  • C End-point RT-PCR analysis of dystrophin mRNA transcripts in CRISPR/Cas9-modified human ⁇ 48-50 DMD myoblasts treated with the indicated sgRNAs. A representative chromatogram of the expected deletion PCR product is shown at the right. Asterisk: band resulting from hybridization of the deletion product strand to the unmodified strand.
  • D Rescue of dystrophin protein expression by CRISPR/Cas9 genome editing was assessed by western blot for the dystrophin protein with GAPDH as a loading control. The arrow indicates the expected restored dystrophin protein band.
  • Fig. 22 shows deletion of the entire exon 45-55 region in human DMD myoblasts by multiplex CRISPR/Cas9 gene editing.
  • A End-point genomic PCR of genomic DNA to detect deletion of the region between intron 44 and intron 55 after treating HEK293Ts or DMD myoblasts with the indicated sgRNAs.
  • B Individual clones of PCR products of the expected size for the deletions from DMD myoblasts in (A) were analyzed by Sanger sequencing to determine the sequences of genomic deletions present at the targeted locus. Below is a representative chromatograms showing the sequence of the expected deletion junctions.
  • C End-point RT-PCR analysis of dystrophin mRNA transcripts in CRISPR/Cas9-modified human ⁇ 48-50 DMD myoblasts treated with the indicated sgRNAs. A representative chromatogram of the expected deletion PCR product is shown at the right.
  • D Analysis of restored dystrophin protein expression by western blot following electroporation of DMD myoblasts with sgRNAs targeted to intron 44 and/or intron 55.
  • Fig. 23 shows verification of flow cytometry-based enrichment of gene-modified DMD myoblasts used for in vivo cell transplantation experiment.
  • DMD myoblasts were treated with Cas9 with or without sgRNA expression vectors for CRl and CR5 and sorted for GFP+ cells by flow cytometry. Deletions at the exon 51 locus were detected by end-point PCR using primers flanking the locus.
  • Neg Ctrl DMD myoblasts treated with Cas9 only and sorted for GFP+ cells.
  • Fig. 24 shows expression of restored human dystrophin in vivo following
  • Fig. 25 shows additional immunofluorescence images probing human dystrophin expression. Serial sections from regions stained with anti-human spectrin are shown inset in top left.
  • A-C Sections from muscles injected with untreated human DMD myoblasts.
  • D-F Sections from muscles injected with CRl/5 treated human DMD myoblasts enriched by flow cytometry. White arrows indicate dystrophin positive fibers.
  • Fig. 26 shows evaluation of CRISPR/Cas9 toxicity and off-target effects for
  • Fig. 27 shows images of TBE-PAGE gels used to quantify Surveyor assay results to measure on-target and off-target gene modification in Table 4. Asterisks mark expected sizes of bands indicative of nuclease activity.
  • Fig. 28 shows end-point nested PCR to detect chromosomal translocations caused by CRISPR/Cas9 off-target activity for CR3 and CR6/CR36 in human cells.
  • Nested end-point PCR analysis was used to detect translocations in (A) HEK293T or sorted hDMD cells treated with Cas9 and CR3 as indicated, (B) HEK293T cells treated with Cas9 and CR36 alone, or (C) sorted hDMD cells treated with Cas9, CR6, and CR36 expression cassettes.
  • the second nested PCR reaction for translocation was amplified using custom primers for each predicted translocation locus to maximize specificity (See Table 4).
  • the schematic depicts the relative location of nested primer pairs used to probe for the presence of translocations.
  • Each possible translocation event was first amplified from genomic DNA isolated from cells treated with or without the indicated sgRNA(s).
  • a second nested PCR reaction was performed using primers within the predicted PCR amplicons that would result from translocations. Expected size was estimated based on the indicated primer binding site and the predicted sgRNA cut site at each locus, ⁇ indicates bands detected at the expected size and verified by Sanger sequencing from each end. indicates amplicons in which Sanger sequencing showed sequences other than the predicted translocation, likely a result of mispriming during the nested PCR.
  • Fig. 29 shows Sanger sequencing chromatograms for bands detected in Fig. 28 resulting from translocations between CR3 and CR3-OT1, on chromosomes X and 1, respectively, in HEK293T cells treated with Cas9 and CR3 gene cassettes. Arrows show regions of homology to the indicated chromosome nearby the expected break points caused by the appropriate sgRNAs. Note that sequencing reads become out of phase near the break point due to the error-prone nature of DNA repair by non-homologous end-joining.
  • Fig. 30 shows Sanger sequencing chromatograms for bands detected in Fig.
  • 26C resulting from translocations between CRl and CR1-OT1, on chromosomes X and 16, respectively, in HEK293T cells treated with Cas9 and CRl gene cassettes. Arrows show regions of homology to the indicated chromosome nearby the expected break points caused by the appropriate sgRNAs. Note that sequencing reads become out of phase near the break point due to the error-prone nature of DNA repair by non-homologous end-joining.
  • Fig. 31 shows an overview of in vivo AAV injections and tissue harvest.
  • Fig. 32 shows Surveyor analysis of Rosa26 ZFN activities in skeletal muscle in vitro and in vivo following delivery of AAV-SASTG-ROSA. Arrows indicate expected bands resulting from Surveyor cleavage, n.d.: not detected, (a) Proliferating C2C12s were transduced with the indicated amount of virus and harvested at 4 days post-infection. Arrows indicate expected bands sizes resulting from Surveyor cleavage, (b) C2C12s were incubated in
  • Fig. 33 shows Rosa T2A opt DNA sequence (SEQ ID NO: 434) and Rosa T2A opt protein sequence (SEQ ID NO: 435).
  • Fig. 34 shows SASTG capsid DNA sequence (SEQ ID NO:436) and SASTG capsid peptide sequence (SEQ ID NO: 437).
  • Fig. 35 shows DZF16 ZFN target site sequence (SEQ ID NO: 442), DZF16-L6 left full amino acid sequence (SEQ ID NO: 443) and DZF16-R6 right full amino acid sequence (SEQ ID NO: 444).
  • Fig. 36 shows E51C3 ZFN target site sequence (SEQ ID NO: 445), E51C3-3L left full amino acid sequence (SEQ ID NO: 446) and E51C3-3R right full amino acid sequence (SEQ ID NO: 447).
  • Fig. 37 shows DZF15 ZFN target site sequence (SEQ ID NO: 448), DZF15-L6 left full amino acid sequence (SEQ ID NO: 449), DZF15-R6 right full amino acid sequence (SEQ ID NO: 450), DZF15-L5 left full amino acid sequence (SEQ ID NO: 451), DZF15-R5 right full amino acid sequence (SEQ ID NO: 452).
  • Fig. 38 shows E51C4 ZFN target site sequence (SEQ ID NO: 453), E51C4-4L left full amino acid sequence (SEQ ID NO: 454) and E51C4-4R right full amino acid sequence (SEQ ID NO: 455).
  • Fig. 39 shows schematic diagrams of a "Single vector, multiplex CRISPR system,” Dual vector, multiplex CRISPR system,” and “Single vector, single gRNA system.”
  • Fig. 40 shows the nucleotide sequences of SaCas9-NLS (with the NLS underlined) (SEQ ID NO: 64) and SaCas9 gRNA (SEQ ID NO: 116).
  • Fig. 41 shows the nucleotide sequences of NmCas9 (with the NLS 1 underlined, the NLS 2 underlined and bolded, and the HA tag bolded), NmCas9 short hairpin from Thomson PNAS 2013 (SEQ ID NO: 118), and NmCas9 long hairpin from Church Nature Biotech 2013 (SEQ ID NO: 119).
  • Fig. 42 shows validation of sgRNA and lentiviral Cas9 expression constructs, (a) Constructs encoding unique Pol III promoters expressing sgRNAs targeting the AAVS 1 locus or a construct containing the hU6 promoter immediately followed by poly-thymidine to terminate expression (“PolyT”) were transfected into HEK293T cells. End-point RT-PCR was used to probe for expression of each indicated promoter/sgRNA construct two days post-transfection.
  • HEK293Ts were transfected with expression vectors encoding the AAVSl zinc-finger nuclease or Cas9-T2A-GFP and the indicated promoter/sgRNA expression cassettes and assessed for gene modification levels 3 days post-transfection using the Surveyor assay
  • HEK293T cells were transduced with lentiviral constructs encoding the indicated Cas9-T2A-GFP constructs without sgRNAs and assessed for Cas9 expression by western blot 7 days post-transduction by probing for a FLAG epitope tag on the N-terminus of the Cas9 protein.
  • Fig. 43 shows Golden Gate assembly of single lentiviral CRISPR/Cas9 expression cassettes.
  • Fig. 44 shows single lentiviral delivery of a multiplex CRISPR/Cas9 system, (a) Four sgRNAs targeting distinct genomic loci were cloned into a lentiviral vector expressing the active Cas9 nuclease, (b) HEK293Ts and primary human dermal fibroblasts were transduced with lentivirus expressing the indicated sgRNAs and assayed for cleavage events using the Surveyor assay. HEK293Ts were assayed 7 days post transduction. The human fibroblasts were assayed 10 days post transduction. [0079] Fig. 45 shows transient gene activation in HEK293Ts stably expressing dCas9-VP64. HEK293Ts were transduced with lentivirus to stably express dCas9-VP64 and were
  • tunable endogenous gene activation of the endogenous IL1RN (a) and HBG1 (b) loci was achieved 3 days post transfection. Peak levels of endogenous IL1RN (c) and HBG1 (d) were observed 3-6 days post transfection and the level of activation returned to background levels between days 15-20. Importantly, the cell lines were able to reactive following a second transfection on day 20 albeit at a lower level than previously observed.
  • Fig. 46 shows stable gene activation in HEK293Ts using a single lentiviral multiplex dCas9-VP64 vector.
  • HEK293Ts were transduced with lentivirus to stably express dCas9-VP64 and the indicated combinations of gRNAs.
  • gRNAs By varying the number of sgRNAs delivered, tunable endogenous gene activation of the endogenous IL1RN (a) and HBG1 (b) loci was achieved
  • Fig. 47 shows IL1RN mRNA expression levels.
  • Fig. 48 shows a schematic representing the direct conversion of fibroblasts to neurons through ectopic expression of the BAM neuronal transcription factors.
  • Fig. 49 shows (A) Schematic of the dCas9-VP64 construct.
  • dCas9-VP64 is a catalytically inactive form of the Cas9 protein fused to a tetramer of the VP 16 transcriptional activation domain.
  • B Schematic showing the mechanism of RNA-guided recruitment of dCas9- VP64 to a genomic target.
  • C Schematic of the experimental protocol to generate iNs with CRISPR/Cas9 transcription factors.
  • Fig. 50 shows endogenous ASCLI expression at day 3 determined by (A) qRT-PCR or total ASCLI protein detected by (B) immunofluorescence in MEFs transduced with dCas9-VP64 and transfected with either gRNAs targeted to the ASCLI promoter, ASCLI cDNA, or luciferase.
  • Asterisk (*) indicates significant (p ⁇ 0.05) increase in ASCLI expression with the co-delivery of
  • Fig. 51 shows (A) TUJ1 and MAP2 -postive cells generated by ectopic BAM factors or by dCas9-VP64 and gRNAs targeted to the BRN2 and ASCLl promoters (B) Cells with neuronal morphology expressing a hSyn-RFP reporter at day 11 in N3 medium.
  • Fig. 52 shows (A) A cell with neuronal morphology positive for the GCaMP5 calcium indicator in the presence (bottom) or absence (top) of KC1 in the culture medium. (B) A trace of normalized fluorescent intensity over time showing depolarization of the cell in response to KC1 addition.
  • Fig. 52 shows activation of downstream targets of Ascll and Brn2, i.e., master regulatory genes, in iCas9-VP64-treated murine embryonic fibroblasts using dCas9-VP64 transcription factors to convert the fibroblasts to neurons.
  • Mouse embryonic fibroblasts (MEFs) were transfected with a control GFP expression plasmid or the iCas9-VP64 expression plasmid and a combination of eight gRNA expression plasmids targeting ASCLl and BRN2.
  • the dCas9 transcription factors were delivered virally. After 10 days in neural induction media, cells were stained for Tuj l, an early marker of neuronal differentiation and MAP2, a marker of more mature neuronal differentiation. The conversion to neurons was efficient.
  • Fig. 53 shows the CRISPR/Cas9 platform for control of mammalian gene regulation.
  • Cas9-based effectors bind genomic sequences in the presence of a chimeric gRNA molecule consisting of a constant region that complexes with Cas9 preceded by an exchangeable 20bp protospacer that confers target site specificity.
  • B. Cas9-based synthetic transcription factors repress transcription of a target gene by interfering with RNA polymerase activity or by binding within the promoter and blocking the binding sites of endogenous transcription factors.
  • Targeting regulatory elements such as enhancers could also potentially block the expression of multiple distal genes.
  • Fig. 54 shows targeting the HS2 enhancer using CRISPR/dCas9-KRAB.
  • the HS2 region is a potent enhancer that distally regulates the expression of globin genes >10kb downstream.
  • a panel of single gRNAs was designed to target sites along the enhancer region.
  • Fig. 55 shows that single gRNAs targeting the HS2 enhancer effect potent
  • A. dCas9 and dCas9-KRAB repressors were delivered on a lentiviral vector. Single gRNAs were transiently transfected for screening. When assayed by quantitative RT-PCR at 3 days post-transfection, K562s expressing dCas9-KRAB achieve up to 80% repression of B. ⁇ -globin, C. ⁇ -globin, and D. ⁇ -globin genes, as compared to control cells that received no gRNA treatment.
  • D. Protein expression in cells expressing dCas9 or dCas9-KRAB and treated with Cr4 or Cr8 show mild repression of ⁇ -globin expression at day 3, compared to ⁇ -actin controls.
  • Fig. 56 shows expression of globin locus genes with varying doses of gRNA plasmid delivered to cells treated with A. no lentivirus, B. dCas9 lentivirus, or C. dCas9-KRAB lentivirus.
  • Increasing the dose of Cr4 gRNA plasmid delivered enhanced repression in dCas9- KRAB treated cells, indicating that both the dCas9-KRAB effector and targeted gRNA play a role in achieving repression.
  • Fig. 57 shows that stably delivering single gRNAs with dCas9-KRAB silences expression of the globin genes.
  • A. dCas9 and dCas9-KRAB repressors were co-expressed on a lentiviral vector with single gRNAs.
  • K562s expressing dCas9-KRAB achieve up to 95% repression of B. ⁇ -globin, C. ⁇ - globin, and D. ⁇ -globin genes, as compared to control cells that received no lentiviral treatment.
  • Fig. 58 shows that isolating the p300 HAT "Core" for targeted epigenetic modification of histones only via dCas9 fusion.
  • Fig. 59 shows a simplified schematic of S. pyogenes dCas9-VP64 fusion (top) and dCas9-p300 core fusion (bottom).
  • the Protospacer Adjacent Motifs (PAM) are shown with arrows at target gene loci and synthetic guide RNA (gRNA) is shown with hatched arrows.
  • Fig. 60A-60C show representative data at three human loci demonstrating the efficacy of activation using dCas9-p300 in relation to dCas9-VP64 and dCas9 without any fused effector domain in the human 293T cell culture line.
  • Fig. 61A-61C show the amino acid sequences of the dCas9 constructs. The legend for all Figs. 61A-61C is shown in Fig. 61 A.
  • Fig. 62 shows that HAT-dCase9-p300 fusion proteins fail to activate gene expression.
  • Fig. 63 shows that gRNA's also act synergistically with dCas9-p300 Core.
  • Fig. 64 shows that dCas9-p300 and dCas9-VP64 do not have any additive effect on transactivation (on different molecules).
  • Fig. 65 shows that dCas9-p300 activates MyoD distal regulation regions, non- synergistically.
  • Fig. 66 shows TALEN mediated integration of minidystrophin at the 5'UTR of the Dp427m skeletal muscle isoform of dystrophin in skeletal myoblast cell lines derived from human DMD patients carrying different deletions in the dystrophin gene.
  • DMD patient cells were electroporated with constructs encoding a TALEN pair active at the 5 'UTR locus and a donor template carrying the minidystrophin gene,
  • Hygromycin-resistant clonal cell lines were isolated and screened by PCR for successful site-specific integrations at the 5'UTR using the primers shown in (a). Asterisks indicate clones selected for further analysis in (c).
  • (c) Clonally isolated DMD myoblasts with detected integration events were differentiated for 6 days and assessed for expression of an HA tag fused to the C terminus of minidystrophin.
  • CRISPR/CRISPR-associated (Cas) 9-based system compositions have been discovered to be useful for altering the expression of genes, genome engineering, and correcting or reducing the effects of mutations in genes involved in genetic diseases.
  • the CRISPR/Cas9-based system involves a Cas9 protein and at least one guide RNA, which provide the DNA targeting specificity for the system.
  • the present disclosure describes a Cas9 fusion protein that combines the DNA sequence targeting function of the CRISPR/Cas9-based system with an additional activity, thus allowing changes in gene expression and/or epigenetic status.
  • the system may also be used in genome engineering and correcting or reducing the effects of gene mutations.
  • the present disclosure also provides certain compositions and methods for delivering CRISPR/CRISPR-associated (Cas) 9-based system and multiple gRNAs to target one or more endogenous genes.
  • Co-transfection of multiple sgRNAs targeted to a single promoter allow for synergistic activation, however, co-transfection of multiple plasmids leads to variable expression levels in each cell due to differences in copy number. Additionally, gene activation following transfection is transient due to dilution of plasmid DNA over time. Moreover, many cell types are not easily transfected and transient gene expression may not be sufficient for inducing a therapeutic effect.
  • a single lentiviral system was developed to express Cas9 and up to four sgRNAs from independent promoters.
  • a platform is disclosed that expresses Cas9 or dCas9 fusion proteins and up to four gRNAs from a single lentiviral vector.
  • the lentiviral vector expresses a constitutive or inducible Cas9 or dCas9-VP64 in addition to one, two, three, or four gRNAs expressed from independent promoters.
  • This system enables control of both the magnitude and timing of CRISPR/Cas9-based gene regulation.
  • the lentiviral platform provides the potent and sustained levels of gene expression that will facilitate therapeutic applications of the CRISPR/Cas9 system in primary cells.
  • this system may be used for editing multiple genes simultaneously, such as the concurrent knockout of several oncogenes.
  • the present disclosure also provides certain compositions and methods for delivering site-specific nucleases to skeletal muscle and cardiac muscle using modified adeno-associated virus (AAV) vectors.
  • the site-specific nucleases which may be engineered, are useful for altering the expression of genes, genome engineering, correcting or reducing the effects of mutations in genes involved in genetic diseases, or manipulating genes involved in other conditions affecting skeletal muscle or cardiac muscle or muscle regeneration.
  • the engineered site-specific nucleases may include a zinc finger nuclease (ZFN), a TAL effector nuclease (TALEN), and/or a CRISPR/Cas9 system for genome editing.
  • ZFN zinc finger nuclease
  • TALEN TAL effector nuclease
  • CRISPR/Cas9 CRISPR/Cas9
  • Gene editing is highly dependent on cell cycle and complex DNA repair pathways that vary from tissue to tissue.
  • Skeletal muscle is a very complex environment, consisting of large myo fibers with more than 100 nuclei per cell.
  • Gene therapy and biologies in general have been limited for decades by in vivo delivery hurdles. These challenges include stability of the carrier in vivo, targeting the right tissue, getting sufficient gene expression and active gene product, and avoiding toxicity that might overcome activity, which is common with gene editing tools.
  • Other delivery vehicles such as direct injection of plasmid DNA, work to express genes in skeletal muscle and cardiac muscle in other contexts, but do not work well with these site-specific nucleases for achieving detectable levels of genome editing.
  • the present disclosure also provides certain fusion epigenetic effector molecules, a dCas9-p300 fusion protein, which provides a robust and potentially more widely applicable tool for synthetic transcriptional modulation compared to the dCas9-VP64 fusion.
  • the activated target genes to a substantially greater extent than the dCas9-VP64 fusion protein at all loci tested.
  • the p300 has intrinsic endogenous activity at enhancers within the human genome.
  • the dCas9-p300 fusion protein may be able to activate endogenous target gene promoters and enhancer regions.
  • the dCas9-p300 fusion protein can be used in human tissue culture cell lines to activate gene expression.
  • This fusion protein may be used to direct the epigenetic state of target loci within human cells with precision and predictability in order to control differentiation, modulate cellular regulation, and apply innovative potential therapies.
  • Current technologies are limited in the strength of activation and the extent and sustainability of epigenetic modulation; obstacles which may be obviated via utilization of this new fusion protein.
  • Adeno-associated virus or "AAV” as used interchangeably herein refers to a small virus belonging to the genus Dependovirus of the Parvoviridae family that infects humans and some other primate species. AAV is not currently known to cause disease and consequently the virus causes a very mild immune response.
  • Binding region refers to the region within a nuclease target region that is recognized and bound by the nuclease.
  • Cardiac muscle or "heart muscle” as used interchangeably herein means a type of involuntary striated muscle found in the walls and histological foundation of the heart, the myocardium. Cardiac muscle is made of cardiomyocytes or myocardiocytes. Myocardiocytes show striations similar to those on skeletal muscle cells but contain only one, unique nucleus, unlike the multinucleated skeletal cells.
  • Cardiac muscle condition refers to a condition related to the cardiac muscle, such as cardiomyopathy, heart failure, arrhythmia, and inflammatory heart disease.
  • Coding sequence or "encoding nucleic acid” as used herein means the nucleic acids (R A or DNA molecule) that comprise a nucleotide sequence which encodes a protein.
  • the coding sequence can further include initiation and termination signals operably linked to regulatory elements including a promoter and polyadenylation signal capable of directing expression in the cells of an individual or mammal to which the nucleic acid is administered.
  • the coding sequence may be codon optimize.
  • “Complement” or “complementary” as used herein means a nucleic acid can mean Watson-Crick (e.g., A-T/U and C-G) or Hoogsteen base pairing between nucleotides or nucleotide analogs of nucleic acid molecules. "Complementarity” refers to a property shared between two nucleic acid sequences, such that when they are aligned antiparallel to each other, the nucleotide bases at each position will be complementary.
  • Correcting or restoring a mutant gene may include replacing the region of the gene that has the mutation or replacing the entire mutant gene with a copy of the gene that does not have the mutation with a repair mechanism such as homology-directed repair (HDR).
  • HDR homology-directed repair
  • Correcting or restoring a mutant gene may also include repairing a frameshift mutation that causes a premature stop codon, an aberrant splice acceptor site or an aberrant splice donor site, by generating a double stranded break in the gene that is then repaired using non-homologous end joining (NHEJ). NHEJ may add or delete at least one base pair during repair which may restore the proper reading frame and eliminate the premature stop codon. Correcting or restoring a mutant gene may also include disrupting an aberrant splice acceptor site or splice donor sequence.
  • NHEJ non-homologous end joining
  • Correcting or restoring a mutant gene may also include deleting a non-essential gene segment by the simultaneous action of two nucleases on the same DNA strand in order to restore the proper reading frame by removing the DNA between the two nuclease target sites and repairing the DNA break by NHEJ.
  • Donor DNA refers to a double-stranded DNA fragment or molecule that includes at least a portion of the gene of interest.
  • the donor DNA may encode a full-functional protein or a partially- functional protein.
  • DMD Duchenne Muscular Dystrophy
  • DMD is a common hereditary monogenic disease and occurs in 1 in 3500 males. DMD is the result of inherited or spontaneous mutations that cause nonsense or frame shift mutations in the dystrophin gene. The majority of dystrophin mutations that cause DMD are deletions of exons that disrupt the reading frame and cause premature translation termination in the dystrophin gene. DMD patients typically lose the ability to physically support themselves during childhood, become progressively weaker during the teenage years, and die in their twenties.
  • Dystrophin refers to a rod-shaped cytoplasmic protein which is a part of a protein complex that connects the cytoskeleton of a muscle fiber to the surrounding extracellular matrix through the cell membrane. Dystrophin provides structural stability to the dystroglycan complex of the cell membrane that is responsible for regulating muscle cell integrity and function.
  • the dystrophin gene or “DMD gene” as used interchangeably herein is 2.2 megabases at locus Xp21. The primary transcription measures about 2,400 kb with the mature mRNA being about 14 kb. 79 exons code for the protein which is over 3500 amino acids.
  • Exon 51 refers to the 51 st exon of the dystrophin gene. Exon 51 is frequently adjacent to frame-disrupting deletions in DMD patients and has been targeted in clinical trials for oligonucleotide-based exon skipping. A clinical trial for the exon 51 skipping compound eteplirsen recently reported a significant functional benefit across 48 weeks, with an average of 47% dystrophin positive fibers compared to baseline. Mutations in exon 51 are ideally suited for permanent correction by NHEJ-based genome editing.
  • “Frameshift” or “frameshift mutation” as used interchangeably herein refers to a type of gene mutation wherein the addition or deletion of one or more nucleotides causes a shift in the reading frame of the codons in the mRNA.
  • the shift in reading frame may lead to the alteration in the amino acid sequence at protein translation, such as a missense mutation or a premature stop codon.
  • a “functional gene” refers to a gene transcribed to mRNA, which is translated to a functional protein.
  • Fusion protein refers to a chimeric protein created through the joining of two or more genes that originally coded for separate proteins. The translation of the fusion gene results in a single polypeptide with functional properties derived from each of the original proteins.
  • Generic construct refers to the DNA or RNA molecules that comprise a nucleotide sequence that encodes a protein.
  • the coding sequence includes initiation and termination signals operably linked to regulatory elements including a promoter and
  • the term "expressible form” refers to gene constructs that contain the necessary regulatory elements operable linked to a coding sequence that encodes a protein such that when present in the cell of the individual, the coding sequence will be expressed.
  • Genetic disease refers to a disease, partially or completely, directly or indirectly, caused by one or more abnormalities in the genome, especially a condition that is present from birth.
  • the abnormality may be a mutation, an insertion or a deletion.
  • the abnormality may affect the coding sequence of the gene or its regulatory sequence.
  • the genetic disease may be, but not limited to DMD, hemophilia, cystic fibrosis, Huntington's chorea, familial hypercholesterolemia (LDL receptor defect), hepatoblastoma, Wilson's disease, congenital hepatic porphyria, inherited disorders of hepatic metabolism, Lesch Nyhan syndrome, sickle cell anemia, thalassaemias, xeroderma pigmentosum, Fanconi's anemia, retinitis pigmentosa, ataxia telangiectasia, Bloom's syndrome, retinoblastoma, and Tay-Sachs disease.
  • DMD hemophilia
  • cystic fibrosis Huntington's chorea
  • hepatoblastoma Wilson's disease
  • congenital hepatic porphyria congenital hepatic porphyria
  • inherited disorders of hepatic metabolism Lesch Nyhan
  • HDR Homology-directed repair
  • HDR uses a donor DNA template to guide repair and may be used to create specific sequence changes to the genome, including the targeted addition of whole genes. If a donor template is provided along with the site specific nuclease, such as with a CRISPR/Cas9-based systems, then the cellular machinery will repair the break by homologous recombination, which is enhanced several orders of magnitude in the presence of DNA cleavage. When the homologous DNA piece is absent, nonhomologous end joining may take place instead.
  • Genome editing refers to changing a gene. Genome editing may include correcting or restoring a mutant gene. Genome editing may include knocking out a gene, such as a mutant gene or a normal gene. Genome editing may be used to treat disease or enhance muscle repair by changing the gene of interest.
  • nucleic acids or polypeptide sequences means that the sequences have a specified percentage of residues that are the same over a specified region. The percentage may be calculated by optimally aligning the two sequences, comparing the two sequences over the specified region, determining the number of positions at which the identical residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the specified region, and multiplying the result by 100 to yield the percentage of sequence identity.
  • the residues of single sequence are included in the denominator but not the numerator of the calculation.
  • thymine (T) and uracil (U) may be considered equivalent.
  • Identity may be performed manually or by using a computer sequence algorithm such as BLAST or BLAST 2.0.
  • mutant gene or “mutated gene” as used interchangeably herein refers to a gene that has undergone a detectable mutation.
  • a mutant gene has undergone a change, such as the loss, gain, or exchange of genetic material, which affects the normal transmission and expression of the gene.
  • a "disrupted gene” as used herein refers to a mutant gene that has a mutation that causes a premature stop codon. The disrupted gene product is truncated relative to a full-length undisrupted gene product.
  • NHEJ Non-homologous end joining pathway
  • the template-independent re-ligation of DNA ends by NHEJ is a stochastic, error-prone repair process that introduces random micro-insertions and micro- deletions (indels) at the DNA breakpoint. This method may be used to intentionally disrupt, delete, or alter the reading frame of targeted gene sequences.
  • NHEJ typically uses short homologous DNA sequences called microhomologies to guide repair. These microhomologies are often present in single-stranded overhangs on the end of double-strand breaks. When the overhangs are perfectly compatible, NHEJ usually repairs the break accurately, yet imprecise repair leading to loss of nucleotides may also occur, but is much more common when the overhangs are not compatible.
  • Normal gene refers to a gene that has not undergone a change, such as a loss, gain, or exchange of genetic material. The normal gene undergoes normal gene transmission and gene expression.
  • Nuclease mediated NHEJ refers to NHEJ that is initiated after a nuclease, such as a cas9, cuts double stranded DNA.
  • nucleic acid or "oligonucleotide” or “polynucleotide” as used herein means at least two nucleotides covalently linked together. The depiction of a single strand also defines the sequence of the complementary strand. Thus, a nucleic acid also encompasses the
  • nucleic acid also encompasses
  • nucleic acids substantially identical nucleic acids and complements thereof.
  • a single strand provides a probe that may hybridize to a target sequence under stringent hybridization conditions.
  • a nucleic acid also encompasses a probe that hybridizes under stringent hybridization conditions.
  • Nucleic acids may be single stranded or double stranded, or may contain portions of both double stranded and single stranded sequence.
  • the nucleic acid may be DNA, both genomic and cDNA, RNA, or a hybrid, where the nucleic acid may contain combinations of deoxyribo- and ribo-nucleotides, and combinations of bases including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine hypoxanthine, isocytosine and isoguanine.
  • Nucleic acids may be obtained by chemical synthesis methods or by recombinant methods.
  • operably linked means that expression of a gene is under the control of a promoter with which it is spatially connected.
  • a promoter may be positioned 5' (upstream) or 3' (downstream) of a gene under its control.
  • the distance between the promoter and a gene may be approximately the same as the distance between that promoter and the gene it controls in the gene from which the promoter is derived. As is known in the art, variation in this distance may be accommodated without loss of promoter function.
  • Partially-functional as used herein describes a protein that is encoded by a mutant gene and has less biological activity than a functional protein but more than a non-functional protein.
  • Premature stop codon or "out-of-frame stop codon” as used interchangeably herein refers to nonsense mutation in a sequence of DNA, which results in a stop codon at location not normally found in the wild-type gene.
  • a premature stop codon may cause a protein to be truncated or shorter compared to the full-length version of the protein.
  • Promoter means a synthetic or naturally-derived molecule which is capable of conferring, activating or enhancing expression of a nucleic acid in a cell.
  • a promoter may comprise one or more specific transcriptional regulatory sequences to further enhance expression and/or to alter the spatial expression and/or temporal expression of same.
  • a promoter may also comprise distal enhancer or repressor elements, which may be located as much as several thousand base pairs from the start site of transcription.
  • a promoter may be derived from sources including viral, bacterial, fungal, plants, insects, and animals.
  • a promoter may regulate the expression of a gene component constitutively, or differentially with respect to cell, the tissue or organ in which expression occurs or, with respect to the developmental stage at which expression occurs, or in response to external stimuli such as physiological stresses, pathogens, metal ions, or inducing agents.
  • promoters include the bacteriophage T7 promoter, bacteriophage T3 promoter, SP6 promoter, lac operator-promoter, tac promoter, SV40 late promoter, SV40 early promoter, RSV-LTR promoter, CMV IE promoter, SV40 early promoter or SV40 late promoter and the CMV IE promoter.
  • RVD Repeat variable diresidue
  • RVD module DNA recognition motif
  • the RVD determines the nucleotide specificity of the RVD module.
  • RVD modules may be combined to produce an RVD array.
  • the "RVD array length” as used herein refers to the number of RVD modules that corresponds to the length of the nucleotide sequence within the TALEN target region that is recognized by a TALEN, i.e., the binding region.
  • Site-specific nuclease refers to an enzyme capable of specifically recognizing and cleaving DNA sequences.
  • the site-specific nuclease may be engineered.
  • engineered site-specific nucleases include zinc finger nucleases (ZFNs), TAL effector nucleases (TALENs), and CRISPR/Cas9-based systems.
  • ZFNs zinc finger nucleases
  • TALENs TAL effector nucleases
  • CRISPR/Cas9-based systems examples include zinc finger nucleases (ZFNs), TAL effector nucleases (TALENs), and CRISPR/Cas9-based systems.
  • Skeletal muscle refers to a type of striated muscle, which is under the control of the somatic nervous system and attached to bones by bundles of collagen fibers known as tendons. Skeletal muscle is made up of individual components known as myocytes, or “muscle cells”, sometimes colloquially called “muscle fibers.” Myocytes are formed from the fusion of developmental myoblasts (a type of embryonic progenitor cell that gives rise to a muscle cell) in a process known as myogenesis. These long, cylindrical, multinucleated cells are also called myo fibers.
  • Sketal muscle condition refers to a condition related to the skeletal muscle, such as muscular dystrophies, aging, muscle degeneration, wound healing, and muscle weakness or atrophy.
  • Spacers and “spacer region” as used interchangeably herein refers to the region within a TALEN or ZFN target region that is between, but not a part of, the binding regions for two TALENs or ZFNs.
  • a mammal ⁇ e.g., cow, pig, camel, llama, horse, goat, rabbit, sheep, hamsters, guinea pig, cat, dog, rat, and mouse
  • a non-human primate for example, a monkey, such as a cynomolgous or rhesus monkey, chimpanzee, etc.
  • the subject may be a human or a non-human.
  • the subject or patient may be
  • Target gene refers to any nucleotide sequence encoding a known or putative gene product.
  • the target gene may be a mutated gene involved in a genetic disease.
  • Target region refers to the region of the target gene to which the site-specific nuclease is designed to bind and cleave.
  • Transcription activator-like effector or “TALE” as used herein refers to a protein structure that recognizes and binds to a particular DNA sequence.
  • TALE DNA-binding domain refers to a DNA-binding domain that includes an array of tandem 33-35 amino acid repeats, also known as RVD modules, each of which specifically recognizes a single base pair of DNA. RVD modules may be arranged in any order to assemble an array that recognizes a defined sequence.
  • a binding specificity of a TALE DNA-binding domain is determined by the RVD array followed by a single truncated repeat of 20 amino acids.
  • a TALE DNA-binding domain may have 12 to 27 RVD modules, each of which contains an RVD and recognizes a single base pair of DNA. Specific RVDs have been identified that recognize each of the four possible DNA nucleotides (A, T, C, and G). Because the TALE DNA-binding domains are modular, repeats that recognize the four different DNA nucleotides may be linked together to recognize any particular DNA sequence. These targeted DNA-binding domains may then be combined with catalytic domains to create functional enzymes, including artificial transcription factors, methyltransferases, integrases, nucleases, and recombinases.
  • TALEN monomer refers to an engineered fusion protein with a catalytic nuclease domain and a designed TALE DNA-binding domain. Two TALEN monomers may be designed to target and cleave a TALEN target region.
  • Transgene refers to a gene or genetic material containing a gene sequence that has been isolated from one organism and is introduced into a different organism. This non-native segment of DNA may retain the ability to produce RNA or protein in the transgenic organism, or it may alter the normal function of the transgenic organism's genetic code. The introduction of a transgene has the potential to change the phenotype of an organism.
  • nucleic acid means (i) a portion or fragment of a referenced nucleotide sequence; (ii) the complement of a referenced nucleotide sequence or portion thereof; (iii) a nucleic acid that is substantially identical to a referenced nucleic acid or the complement thereof; or (iv) a nucleic acid that hybridizes under stringent conditions to the referenced nucleic acid, complement thereof, or a sequences substantially identical thereto.
  • Variant with respect to a peptide or polypeptide that differs in amino acid sequence by the insertion, deletion, or conservative substitution of amino acids, but retain at least one biological activity.
  • Variant may also mean a protein with an amino acid sequence that is substantially identical to a referenced protein with an amino acid sequence that retains at least one biological activity.
  • a conservative substitution of an amino acid i.e., replacing an amino acid with a different amino acid of similar properties ⁇ e.g. , hydrophilicity, degree and distribution of charged regions
  • minor changes may be identified, in part, by considering the hydropathic index of amino acids, as understood in the art. Kyte et al., J. Mol. Biol.
  • the hydropathic index of an amino acid is based on a consideration of its hydrophobicity and charge. It is known in the art that amino acids of similar hydropathic indexes may be substituted and still retain protein function. In one aspect, amino acids having hydropathic indexes of ⁇ 2 are substituted.
  • the hydrophilicity of amino acids may also be used to reveal substitutions that would result in proteins retaining biological function. A consideration of the hydrophilicity of amino acids in the context of a peptide permits calculation of the greatest local average hydrophilicity of that peptide. Substitutions may be performed with amino acids having hydrophilicity values within ⁇ 2 of each other.
  • hydrophobicity index and the hydrophilicity value of amino acids are influenced by the particular side chain of that amino acid. Consistent with that observation, amino acid substitutions that are compatible with biological function are understood to depend on the relative similarity of the amino acids, and particularly the side chains of those amino acids, as revealed by the hydrophobicity, hydrophilicity, charge, size, and other properties.
  • Vector as used herein means a nucleic acid sequence containing an origin of replication.
  • a vector may be a viral vector, bacteriophage, bacterial artificial chromosome or yeast artificial chromosome.
  • a vector may be a DNA or R A vector.
  • a vector may be a self- replicating extrachromosomal vector, and preferably, is a DNA plasmid.
  • the vector may encode an iCas9-VP64 fusion protein comprising the amino acid sequence of SEQ ID NO: 1 or at least one gRNA nucleotide sequence of any one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, and 585-625.
  • the vector may encode Cas9 and at least one gRNA nucleotide sequence of any one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, and 585-625.
  • Zinc finger refers to a protein structure that recognizes and binds to DNA sequences.
  • the zinc finger domain is the most common DNA-binding motif in the human proteome.
  • a single zinc finger contains approximately 30 amino acids and the domain typically functions by binding 3 consecutive base pairs of DNA via interactions of a single amino acid side chain per base pair.
  • Zinc finger nuclease or "ZFN” as used interchangeably herein refers to a chimeric protein molecule comprising at least one zinc finger DNA binding domain effectively linked to at least one nuclease or part of a nuclease capable of cleaving DNA when fully assembled.
  • compositions for genome editing, genomic alteration or altering gene expression of a target gene may include a may include viral vector and fusion protein such as a site-specific nuclease or CRISPR/Cas9-system with at least one gRNA.
  • the present invention is directed to a composition for genome editing a target gene in skeletal muscle or cardiac muscle of a subject.
  • the composition includes a modified AAV vector and a nucleotide sequence encoding a site-specific nuclease.
  • the composition delivers active forms of site-specific nucleases to skeletal muscle or cardiac muscle.
  • the composition may further comprise a donor DNA or a transgene.
  • the target gene may be involved in differentiation of a cell or any other process in which activation, repression, or disruption of a gene may be desired, or may have a mutation such as a deletion, frameshift mutation, or a nonsense mutation. If the target gene has a mutation that causes a premature stop codon, an aberrant splice acceptor site or an aberrant splice donor site, the site-specific nucleases may be designed to recognize and bind a nucleotide sequence upstream or downstream from the premature stop codon, the aberrant splice acceptor site or the aberrant splice donor site.
  • the site-specific nucleases may also be used to disrupt normal gene splicing by targeting splice acceptors and donors to induce skipping of premature stop codons or restore a disrupted reading frame.
  • the site-specific nucleases may or may not mediate off-target changes to protein-coding regions of the genome.
  • CRISPRs refers to loci containing multiple short direct repeats that are found in the genomes of approximately 40% of sequenced bacteria and 90% of sequenced archaea.
  • the CRISPR system is a microbial nuclease system involved in defense against invading phages and plasmids that provides a form of acquired immunity.
  • the CRISPR loci in microbial hosts contain a combination of CRISPR-associated (Cas) genes as well as non-coding RNA elements capable of programming the specificity of the CRISPR-mediated nucleic acid cleavage.
  • Spacers Short segments of foreign DNA, called spacers, are incorporated into the genome between CRISPR repeats, and serve as a 'memory' of past exposures.
  • Cas9 forms a complex with the 3' end of the sgRNA, and the protein-RNA pair recognizes its genomic target by complementary base pairing between the 5' end of the sgRNA sequence and a predefined 20 bp DNA sequence, known as the protospacer.
  • This complex is directed to homologous loci of pathogen DNA via regions encoded within the crRNA, i.e., the protospacers, and protospacer-adjacent motifs (PAMs) within the pathogen genome.
  • PAMs protospacer-adjacent motifs
  • the non-coding CRISPR array is transcribed and cleaved within direct repeats into short crRNAs containing individual spacer sequences, which direct Cas nucleases to the target site (protospacer).
  • the Cas9 nuclease can be directed to new genomic targets.
  • CRISPR spacers are used to recognize and silence exogenous genetic elements in a manner analogous to RNAi in eukaryotic organisms.
  • Type II effector system carries out targeted DNA double-strand break in four sequential steps, using a single effector enzyme, Cas9, to cleave dsDNA.
  • Cas9 effector enzyme
  • the Type II effector system may function in alternative contexts such as eukaryotic cells.
  • the Type II effector system consists of a long pre-crRNA, which is transcribed from the spacer-containing CRISPR locus, the Cas9 protein, and a tracrRNA, which is involved in pre-crRNA processing.
  • the tracrRNAs hybridize to the repeat regions separating the spacers of the pre-crRNA, thus initiating dsRNA cleavage by endogenous RNase III. This cleavage is followed by a second cleavage event within each spacer by Cas9, producing mature crRNAs that remain associated with the tracrRNA and Cas9, forming a Cas9:crRNA-tracrRNA complex.
  • the Cas9:crRNA-tracrRNA complex unwinds the DNA duplex and searches for sequences matching the crRNA to cleave.
  • Target recognition occurs upon detection of complementarity between a "protospacer" sequence in the target DNA and the remaining spacer sequence in the crRNA.
  • Cas9 mediates cleavage of target DNA if a correct protospacer-adjacent motif (PAM) is also present at the 3 ' end of the protospacer.
  • PAM protospacer-adjacent motif
  • the sequence must be immediately followed by the protospacer-adjacent motif (PAM), a short sequence recognized by the Cas9 nuclease that is required for DNA cleavage.
  • PAM protospacer-adjacent motif
  • Different Type II systems have differing PAM requirements. The S.
  • pyogenes CRISPR system may have the PAM sequence for this Cas9 (SpCas9) as 5'-NRG-3', where R is either A or G, and characterized the specificity of this system in human cells.
  • SpCas9 the PAM sequence for this Cas9
  • a unique capability of the CRISPR/Cas9 system is the straightforward ability to simultaneously target multiple distinct genomic loci by co-expressing a single Cas9 protein with two or more sgRNAs.
  • the Streptococcus pyogenes Type II system naturally prefers to use an "NGG” sequence, where "N” can be any nucleotide, but also accepts other PAM sequences, such as "NAG” in engineered systems (Hsu et al, Nature Biotechnology (2013) doi: 10.1038/nbt.2647).
  • NmCas9 derived from Neisseria meningitidis
  • NmCas9 normally has a native PAM of NNNNGATT, but has activity across a variety of PAMs, including a highly degenerate NNNNGNNN PAM (Esvelt et al. Nature Methods (2013) doi: 10.1038/nmeth.2681).
  • the CRISPR/Cas9-based engineered systems may be designed to target any gene, including genes involved in a genetic disease, aging, tissue regeneration, or wound healing.
  • the CRISPR/Cas9-based systems may include a Cas9 protein or Cas9 fusion protein and at least one gRNA.
  • the Cas9 fusion protein may, for example, include a domain that has a different activity that what is endogenous to Cas9, such as a transactivation domain.
  • the target gene may be involved in differentiation of a cell or any other process in which activation of a gene may be desired, or may have a mutation such as a frameshift mutation or a nonsense mutation. If the target gene has a mutation that causes a premature stop codon, an aberrant splice acceptor site or an aberrant splice donor site, the CRISPR/Cas9-based system may be designed to recognize and bind a nucleotide sequence upstream or downstream from the premature stop codon, the aberrant splice acceptor site or the aberrant splice donor site.
  • the CRISPR-Cas9-based system may also be used to disrupt normal gene splicing by targeting splice acceptors and donors to induce skipping of premature stop codons or restore a disrupted reading frame.
  • the CRISPR/Cas9-based system may or may not mediate off-target changes to protein- coding regions of the genome,
  • the CRISPR/Cas9-based system may include a Cas9 protein or a Cas9 fusion protein.
  • Cas9 protein is an endonuclease that cleaves nucleic acid and is encoded by the CRISPR loci and is involved in the Type II CRISPR system.
  • the Cas9 protein may be from any bacterial or archaea species, such as Streptococcus pyogenes.
  • the Cas9 protein may be mutated so that the nuclease activity is inactivated.
  • iCas9 an inactivated Cas9 protein from Streptococcus pyogenes (iCas9, also referred to as “dCas9") with no endonuclease activity has been recently targeted to genes in bacteria, yeast, and human cells by gR As to silence gene expression through steric hindrance.
  • iCas9 and dCas9 both refer to a Cas9 protein that has the amino acid substitutions D10A and H840A and has its nuclease activity inactivated.
  • the CRISPR/Cas9-based system may include a Cas9 of SEQ ID NO: 459 or 461.
  • the CRISPR/Cas9-based system may include a fusion protein.
  • the fusion protein may comprise two heterologous polypeptide domains, wherein the first polypeptide domain comprises a Cas protein and the second polypeptide domain has an activity such as transcription activation activity, transcription repression activity, transcription release factor activity, histone modification activity, nuclease activity, nucleic acid association activity, methylase activity, or demethylase activity.
  • the fusion protein may include a Cas9 protein or a mutated Cas9 protein, as described above, fused to a second polypeptide domain that has an activity such as transcription activation activity, transcription repression activity, transcription release factor activity, histone modification activity, nuclease activity, nucleic acid association activity, methylase activity, or demethylase activity.
  • the second polypeptide domain may have transcription activation activity, i.e., a transactivation domain.
  • a transactivation domain For example, gene expression of endogenous mammalian genes, such as human genes, may be achieved by targeting a fusion protein of iCas9 and a transactivation domain to mammalian promoters via combinations of gRNAs.
  • the transactivation domain may include a VP 16 protein, multiple VP 16 proteins, such as a VP48 domain or VP64 domain, or p65 domain of NF kappa B transcription activator activity.
  • the fusion protein may be iCas9-VP64.
  • the second polypeptide domain may have transcription repression activity.
  • the second polypeptide domain may have a Kruppel associated box activity, such as a KRAB domain, ERF repressor domain activity, Mxil repressor domain activity, SID4X repressor domain activity, Mad-SID repressor domain activity or TATA box binding protein activity.
  • the fusion protein may be dCas9-KRAB.
  • the second polypeptide domain may have transcription release factor activity.
  • the second polypeptide domain may have eukaryotic release factor 1 (ERF1) activity or eukaryotic release factor 3 (ERF3) activity.
  • the second polypeptide domain may have histone modification activity.
  • the second polypeptide domain may have histone deacetylase, histone acetyltransferase, histone
  • the histone acetyltransferase may be p300 or CREB-binding protein (CBP) protein, or fragments thereof.
  • CBP CREB-binding protein
  • the fusion protein may be dCas9-p300.
  • the second polypeptide domain may have nuclease activity that is different from the nuclease activity of the Cas9 protein.
  • a nuclease, or a protein having nuclease activity is an enzyme capable of cleaving the phosphodiester bonds between the nucleotide subunits of nucleic acids.
  • Nucleases are usually further divided into endonucleases and exonucleases, although some of the enzymes may fall in both categories.
  • Well known nucleases are deoxyribonuclease and ribonuclease.
  • the second polypeptide domain may have nucleic acid association activity or nucleic acid binding protein- DNA-binding domain (DBD) is an independently folded protein domain that contains at least one motif that recognizes double- or single-stranded DNA.
  • a DBD can recognize a specific DNA sequence (a recognition sequence) or have a general affinity to DNA.
  • nucleic acid association region selected from the group consisting of helix-turn-helix region, leucine zipper region, winged helix region, winged helix-turn-helix region, helix-loop-helix region, immunoglobulin fold, B3 domain, Zinc finger, HMG-box, Wor3 domain, TAL effector DNA-binding domain.
  • the second polypeptide domain may have methylase activity, which involves transferring a methyl group to DNA, RNA, protein, small molecule, cytosine or adenine.
  • the second polypeptide domain may include a DNA methyltransferase.
  • the second polypeptide domain may have demethylase activity.
  • the second polypeptide domain may include an enzyme that remove methyl (CH3-) groups from nucleic acids, proteins (in particular histones), and other molecules.
  • the second polypeptide domain may include an enzyme that remove methyl (CH3-) groups from nucleic acids, proteins (in particular histones), and other molecules.
  • polypeptide may covert the methyl group to hydroxymethylcytosine in a mechanism for demethylating DNA.
  • the second polypeptide may catalyze this reaction.
  • the second polypeptide that catalyzes this reaction may be Tetl .
  • the gRNA provides the targeting of the CRISPR/Cas9-based system.
  • the gRNA is a fusion of two noncoding RNAs: a crRNA and a tracrRNA.
  • the sgRNA may target any desired DNA sequence by exchanging the sequence encoding a 20 bp protospacer which confers targeting specificity through complementary base pairing with the desired DNA target.
  • gRNA mimics the naturally occurring crRNA:tracrRNA duplex involved in the Type II Effector system. This duplex, which may include, for example, a 42-nucleotide crRNA and a 75 -nucleotide tracrRNA, acts as a guide for the Cas9 to cleave the target nucleic acid.
  • target region refers to the region of the target gene to which the CRISPR/Cas9-based system targets.
  • the CRISPR/Cas9-based system may include at least one gRNA, wherein the gRNAs target different DNA sequences.
  • the target DNA sequences may be overlapping.
  • the target sequence or protospacer is followed by a PAM sequence at the 3 ' end of the protospacer.
  • Different Type II systems have differing PAM requirements.
  • the Streptococcus pyogenes Type II system uses an "NGG" sequence, where "N" can be any nucleotide.
  • the number of gRNA administered to the cell may be at least 1 gRNA, at least 2 different gRNA, at least 3 different gRNA at least 4 different gRNA, at least 5 different gRNA, at least 6 different gRNA, at least 7 different gRNA, at least 8 different gRNA, at least 9 different gRNA, at least 10 different gRNAs, at least 11 different gRNAs, at least 12 different gRNAs, at least 13 different gRNAs, at least 14 different gRNAs, at least 15 different gRNAs, at least 16 different gRNAs, at least 17 different gRNAs, at least 18 different gRNAs, at least 18 different gRNAs, at least 20 different gRNAs, at least 25 different gRNAs, at least 30 different gRNAs, at least 35 different gRNAs, at least 40 different gRNAs, at least 45 different gRNAs, or at least 50 different gRNAs.
  • the number of gRNA administered to the cell may be between at least 1 gRNA to at least 50 different gRNAs, at least 1 gRNA to at least 45 different gRNAs, at least 1 gRNA to at least 40 different gRNAs, at least 1 gRNA to at least 35 different gRNAs, at least 1 gRNA to at least 30 different gRNAs, at least 1 gRNA to at least 25 different gRNAs, at least 1 gRNA to at least 20 different gRNAs, at least 1 gRNA to at least 16 different gRNAs, at least 1 gRNA to at least 12 different gRNAs, at least 1 gRNA to at least 8 different gRNAs, at least 1 gRNA to at least 4 different gRNAs, at least 4 gRNAs to at least 50 different gRNAs, at least 4 different gRNAs to at least 45 different gRNAs, at least 4 different gRNAs to at least 40 different gRNAs, at least 4 different gRNAs to at least 35
  • the gRNA may comprise a complementary polynucleotide sequence of the target DNA sequence followed by a PAM sequence.
  • the gRNA may comprise a "G" at the 5' end of the complementary polynucleotide sequence.
  • the gRNA may comprise at least a 10 base pair, at least a l l base pair, at least a 12 base pair, at least a 13 base pair, at least a 14 base pair, at least a 15 base pair, at least a 16 base pair, at least a 17 base pair, at least a 18 base pair, at least a 19 base pair, at least a 20 base pair, at least a 21 base pair, at least a 22 base pair, at least a 23 base pair, at least a 24 base pair, at least a 25 base pair, at least a 30 base pair, or at least a 35 base pair complementary polynucleotide sequence of the target DNA sequence followed by a PAM sequence.
  • the PAM sequence may be "NGG", where "N” can be any nucleotide.
  • the gRNA may target at least one of the promoter region, the enhancer region or the transcribed region of the target gene.
  • the gRNA may include a nucleic acid sequence of at least one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, 585-625, 462 (Fig. 40), 464 (Fig. 41), and 465 (Fig. 41).
  • the gRNA may target any nucleic acid sequence.
  • the nucleic acid sequence target may be DNA.
  • the DNA may be any gene.
  • the gRNA may target a gene, such as BRN2, MYT1L, ASCII, NANOG, VEGFA, TERT, IL1B, IL1R2, IL1RN, HBG1, HBG2, MYOD1, OCT4, and DMD.
  • Dystrophin is a rod- shaped cytoplasmic protein which is a part of a protein complex that connects the cytoskeleton of a muscle fiber to the surrounding extracellular matrix through the cell membrane.
  • Dystrophin provides structural stability to the dystroglycan complex of the cell membrane.
  • the dystrophin gene is 2.2 megabases at locus Xp21.
  • the primary transcription measures about 2,400 kb with the mature m NA being about 14 kb.
  • Normal skeleton muscle tissue contains only small amounts of dystrophin but its absence of abnormal expression leads to the development of severe and incurable symptoms.
  • dystrophin gene Some mutations in the dystrophin gene lead to the production of defective dystrophin and severe dystrophic phenotype in affected patients. Some mutations in the dystrophin gene lead to partially-functional dystrophin protein and a much milder dystrophic phenotype in affected patients.
  • DMD is the result of inherited or spontaneous mutations that cause nonsense or frame shift mutations in the dystrophin gene.
  • Naturally occurring mutations and their consequences are relatively well understood for DMD. It is known that in-frame deletions that occur in the exon 45-55 region contained within the rod domain can produce highly functional dystrophin proteins, and many carriers are asymptomatic or display mild symptoms. Furthermore, more than 60% of patients may theoretically be treated by targeting exons in this region of the dystrophin gene. Efforts have been made to restore the disrupted dystrophin reading frame in DMD patients by skipping non-essential exons during mRNA splicing to produce internally deleted but functional dystrophin proteins. The deletion of internal dystrophin exons retain the proper reading frame but cause the less severe Becker muscular dystrophy.
  • a CRISPR/Cas9-based system specific for dystrophin gene are disclosed herein.
  • the CRISPR/Cas9-based system may include Cas9 and at least one gRNA to target the dystrophin gene.
  • the CRISPR/Cas9-based system may bind and recognize a target region.
  • the target regions may be chosen immediately upstream of possible out-of- frame stop codons such that insertions or deletions during the repair process restore the dystrophin reading frame by frame conversion.
  • Target regions may also be splice acceptor sites or splice donor sites, such that insertions or deletions during the repair process disrupt splicing and restore the dystrophin reading frame by splice site disruption and exon exclusion.
  • Target regions may also be aberrant stop codons such that insertions or deletions during the repair process restore the dystrophin reading frame by eliminating or disrupting the stop codon.
  • Single or multiplexed sgRNAs may be designed to restore the dystrophin reading frame by targeting the mutational hotspot at exons 45-55 and introducing either intraexonic small insertions and deletions, or large deletions of one or more exons.
  • dystrophin expression may be restored in Duchenne patient muscle cells in vitro. Human dystrophin was detected in vivo following transplantation of genetically corrected patient cells into immunodeficient mice.
  • the unique multiplex gene editing capabilities of the CRISPR/Cas9 system enable efficiently generating large deletions of this mutational hotspot region that can correct up to 62% of patient mutations by universal or patient- specific gene editing approaches.
  • the CRISPR/Cas9-based system may use gRNA of varying sequences and lengths. Examples of gRNAs may be found in Table 6.
  • the CRISPR/Cas9-based system may target a nucleic acid sequence of SEQ ID NOs: 65-144, or a complement thereof.
  • the gRNA may include a nucleotide sequence selected from the group consisting of SEQ ID NO: 65-144, or a complement thereof.
  • the disclosed CRISPR/Cas9-based systems were engineered to mediate highly efficient gene editing at exon 51 of the dystrophin gene. These CRISPR/Cas9- based systems restored dystrophin protein expression in cells from DMD patients.
  • Exon 51 is frequently adjacent to frame-disrupting deletions in DMD. Elimination of exon 51 from the dystrophin transcript by exon skipping can be used to treat approximately 15% of all DMD patients.
  • This class of DMD mutations is ideally suited for permanent correction by NHEJ-based genome editing and HDR.
  • the CRISPR/Cas9-based systems described herein have been developed for targeted modification of exon 51 in the human dystrophin gene. These CRISPR/Cas9-based systems were transfected into human DMD cells and mediated efficient gene modification and conversion to the correct reading frame. Protein restoration was concomitant with frame restoration and detected in a bulk population of CRISPR/Cas9-based system -treated cells. Similarly, the elimination of exons 45-55 of the dystrophin transcript can be used to treat approximately 62% of all DMD patients. (3) AAV/CRISPR Constructs
  • AAV may be used to deliver CRISPRs using various construct configurations (see Fig. 39).
  • AAV may deliver Cas9 and gRNA expression cassettes on separate vectors.
  • the small Cas9 proteins derived from species such as Staphylococcus aureus or Neisseria meningitidis, are used then both the Cas9 and up to two gRNA expression cassettes may be combined in a single AAV vector within the 4.7 kb packaging limit (see Fig. 39).
  • the present disclosure is directed to a multiplex CRISPR/Cas9-Based System which includes a CRISPR/CRISPR-associated (Cas) 9-based system, such as Cas9 or dCas9, and multiple gRNAs to target one or more endogenous genes.
  • a CRISPR/CRISPR-associated (Cas) 9-based system such as Cas9 or dCas9
  • This platform utilizes a convenient Golden Gate cloning method to rapidly incorporate up to four independent sgRNA expression cassettes into a single lentiviral vector.
  • Each sgRNA was efficiently expressed and could mediate multiplex gene editing at diverse loci in immortalized and primary human cell lines.
  • Transient transcriptional activation in cell lines stably expressing dCas9-VP64 was demonstrated to be tunable by synergistic activation with one to four sgRNAs.
  • the single lentiviral vector can induce sustained and long-term endogenous gene expression in immortalized and primary
  • the multiplex CRISPR/Cas9-Based System provides potency of transcriptional activation and tunable induction of transcriptional activation.
  • the final vector expresses a constitutive Cas9 or dCas9-VP64 in addition to one, two, three, or four sgRNAs expressed from independent promoters.
  • Each promoter is capable of efficiently expressing sgRNAs that direct similar levels of Cas9 nuclease activity.
  • lentiviral delivery of a single vector expressing Cas9 and four sgRNAs targeting independent loci resulted in simultaneous multiplex gene editing of all four loci.
  • the multiplex CRISPR/Cas9-Based System allows efficient multiplex gene editing for simultaneously inactivating multiple genes.
  • the CRISPR/Cas9 system can simultaneously target multiple distinct genomic loci by co-expressing a single Cas9 protein with two or more sgRNAs, making this system uniquely suited for multiplex gene editing or synergistic activation applications.
  • the CRISPR/Cas9 system greatly expedites the process of molecular targeting to new sites by simply modifying the expressed sgRNA molecule.
  • the single ientiviral vector may ⁇ be combined with methods for achieving inducible control of these components, either by chemical or optogenetic regulation, to facilitate investigation of the dynamics of gene regulation in both time and space.
  • the multiplex CRISPR/Cas9-based systems may transcriptionally activate two or more endogenous genes.
  • the multiplex CRISPR/Cas9-based systems may transcriptionally repress two or more endogenous genes.
  • at least two endogenous genes, at least three endogenous genes, at least four endogenous genes, at least five endogenous genes, or at least ten endogenous genes may be activated or repressed by the multiplex CRISPR/Cas9-based system.
  • Between two and fifteen genes, between two and ten genes, between two and five genes, between five and fifteen genes, or between five and ten genes may be activated or repressed by the multiplex CRISPR/Cas9-based system.
  • the multiplex CRISPR/Cas9-based system includes a modified lentiviral vector.
  • the modified lentiviral vector includes a first polynucleotide sequence encoding a fusion protein and a second polynucleotide sequence encoding at least one sgRNA.
  • the fusion protein may be the fusion protein of the CRISPR/Cas9-based system, as described above.
  • the first polynucleotide sequence may be operably linked to a promoter.
  • the promoter may be a constitutive promoter, an inducible promoter, a repressible promoter, or a regulatable promoter.
  • the second polynucleotide sequence encodes at least 1 sgRNA.
  • the second polynucleotide sequence may encode at least 1 sgRNA, at least 2 sgRNAs, at least 3 sgRNAs, at least 4 sgRNAs, at least 5 sgRNAs, at least 6 sgRNAs, at least 7 sgRNAs, at least 8 sgRNAs, at least 9 sgRNAs, at least 10 sgRNAs, at least 11 sgRNA, at least 12 sgRNAs, at least 13 sgRNAs, at least 14 sgRNAs, at least 15 sgRNAs, at least 16 sgRNAs, at least 17 sgRNAs, at least 18 sgRNAs, at least 19 sgRNAs, at least 20 sgRNAs, at least 25 sgRNA, at least 30 sgRNAs, at least 35 sgRNAs, at least 40 sgRNAs, at least 45
  • the second polynucleotide sequence may encode between 1 sgRNA and 50 sgRNAs, between 1 sgRNA and 45 sgRNAs, between 1 sgRNA and 40 sgRNAs, between 1 sgRNA and 35 sgRNAs, between 1 sgRNA and 30 sgRNAs, between 1 sgRNA and 25 different sgRNAs, between 1 sgRNA and 20 sgRNAs, between 1 sgRNA and 16 sgRNAs, between 1 sgRNA and 8 different sgRNAs, between 4 different sgRNAs and 50 different sgRNAs, between 4 different sgRNAs and 45 different sgRNAs, between 4 different sgRNAs and 40 different sgRNAs, between 4 different sgRNAs and 35 different sgRNAs, between 4 different sgRNAs and 30 different sgRNAs, between 4 different sgRNAs and 25 different sgRNAs, between 4 different sgRNAs and 20 different
  • Each of the polynucleotide sequences encoding the different sgRNAs may be operably linked to a promoter.
  • the promoters that are operably linked to the different sgRNAs may be the same promoter.
  • the promoters that are operably linked to the different sgRNAs may be different promoters.
  • the promoter may be a constitutive promoter, an inducible promoter, a repressible promoter, or a regulatable promoter.
  • the at least one sgRNA may bind to a target gene or loci. If more than one sgRNA is included, each of the sgRNAs binds to a different target region within one target loci or each of the sgRNA binds to a different target region within different gene loci.
  • the fusion protein may include Cas9 protein or iCas9-VP64 protein.
  • the fusion protein may include a VP64 domain, a p300 domain, or a KRAB domain.
  • the composition includes a nucleotide sequence encoding a site- specific nuclease that binds and cleaves a target region.
  • the site-specific nuclease may be engineered.
  • an engineered site-specific nuclease may be a CRISPR/Cas9-based system, a ZFN, or a TALEN.
  • the site-specific nuclease may bind and cleave a gene or locus in the genome of a cell in the skeletal muscle or cardiac muscle.
  • the gene or locus may be the Rosa26 locus or the dystrophin gene.
  • the CRISPR/Cas9-based system may be used to introduce site- specific double strand breaks at targeted genomic loci.
  • the site-specific nuclease may be a ZFN.
  • a single zinc finger contains approximately 30 amino acids and the domain functions by binding 3 consecutive base pairs of DNA via interactions of a single amino acid side chain per base pair.
  • the modular structure of the zinc finger motif permits the conjunction of several domains in series, allowing for the recognition and targeting of extended sequences in multiples of 3 nucleotides.
  • These targeted DNA-binding domains can be combined with a nuclease domain, such as Fokl, to generate a site-specific nuclease, called a "zinc finger nuclease" (ZFNs) that can be used to introduce site-specific double strand breaks at targeted genomic loci.
  • ZFNs zinc finger nuclease
  • the ZFN may target the Rosa26 locus (Perez-Pinera et al. Nucleic Acids Research (2012) 40:3741-3752) or a dystrophin gene. Examples of ZFNs are shown in Table 1 and Figs. 35-38. In Table 1, the DNA recognition helices are underlined and "Fok ELD-S” and “Fok KKR-S” refers to the Fokl nuclease domain that is fused to the zinc finger protein DNA-binding domains. In Figs.
  • the target DNA sequence in the target sites i.e., in SEQ ID NOs: 442, 445, 448, and 453
  • the DNA recognition helices in the ZFN amino acid sequences i.e., in SEQ ID NOs: 443, 444, 446, 447, 449-452, 454, and 455 are underlined, respectively.
  • TALENs may be used to introduce site-specific double strand breaks at targeted genomic loci. Site-specific double-strand breaks are created when two independent TALENs bind to nearby DNA sequences, thereby permitting dimerization of Fokl and cleavage of the target DNA. TALENs have advanced genome editing due to their high rate of successful and efficient genetic modification. This DNA cleavage may stimulate the natural DNA-repair machinery, leading to one of two possible repair pathways: homo logy-directed repair (HDR) or the non-homologous end joining (NHEJ) pathway.
  • HDR homo logy-directed repair
  • NHEJ non-homologous end joining
  • the TALENs may be designed to target any gene involved in a genetic disease.
  • the TALENs may include a nuclease and a TALE DNA-binding domain that binds to the target gene in a TALEN target region.
  • the target gene may have a mutation such as a frameshift mutation or a nonsense mutation. If the target gene has a mutation that causes a premature stop codon, the TALEN may be designed to recognize and bind a nucleotide sequence upstream or downstream from the premature stop codon.
  • a "TALEN target region” includes the binding regions for two TALENs and the spacer region, which occurs between the binding regions. The two TALENs bind to different binding regions within the TALEN target region, after which the TALEN target region is cleaved. Examples of TALENs are described in International Patent Application No. PCT/US2013/038536, which is incorporated by reference in its entirety.
  • the composition includes a nucleotide sequence encoding a transcriptional activator that activates a target gene.
  • the transcriptional activator may be engineered.
  • an engineered transcriptional activator may be a CRISPR/Cas9-based system, a zinc finger fusion protein, or a TALE fusion protein.
  • the CRISPR/Cas9-based system may be used to activate transcription of a target gene with RNA.
  • the CRISPR/Cas9-based system may include a fusion protein, as described above, wherein the second polypeptide domain has transcription activation activity or histone modification activity.
  • the second polypeptide domain may include VP64 or p300.
  • the transcriptional activator may be a zinc finger fusion protein.
  • the zinc finger targeted DNA-binding domains as described above, can be combined with a domain that has transcription activation activity or histone modification activity.
  • the domain may include VP64 or p300.
  • TALE fusion proteins may be used to activate transcription of a target gene.
  • the TALE fusion protein may include a TALE DNA-binding domain and a domain that has transcription activation activity or histone modification activity.
  • the domain may include VP64 or p300.
  • the present invention is directed to a composition for altering gene expression and engineering or altering genomic DNA in a cell or subject.
  • the composition may also include a viral delivery system.
  • the present invention is directed to a composition for genome editing a target gene in skeletal muscle or cardiac muscle of a subject.
  • the composition includes a modified AAV vector and a nucleotide sequence encoding a site-specific nuclease.
  • the composition delivers active forms of site-specific nucleases to skeletal muscle or cardiac muscle.
  • the composition may further comprise a donor DNA or a transgene.
  • the target gene may be involved in differentiation of a cell or any other process in which activation, repression, or disruption of a gene may be desired, or may have a mutation such as a deletion, frameshift mutation, or a nonsense mutation. If the target gene has a mutation that causes a premature stop codon, an aberrant splice acceptor site or an aberrant splice donor site, the site-specific nucleases may be designed to recognize and bind a nucleotide sequence upstream or downstream from the premature stop codon, the aberrant splice acceptor site or the aberrant splice donor site.
  • the site-specific nucleases may also be used to disrupt normal gene splicing by targeting splice acceptors and donors to induce skipping of premature stop codons or restore a disrupted reading frame.
  • the site-specific nucleases may or may not mediate off-target changes to protein-coding regions of the genome.
  • the composition includes a modified adeno-associated virus (AAV) vector.
  • the modified AAV vector may have enhanced cardiac and skeletal muscle tissue tropism.
  • the modified AAV vector may be capable of delivering and expressing the site- specific nuclease in the cell of a mammal.
  • the modified AAV vector may be an AAV-SASTG vector (Piacentino et al. (2012) Human Gene Therapy 23:635-646).
  • the modified AAV vector may deliver nucleases to skeletal and cardiac muscle in vivo.
  • the modified AAV vector may be based on one or more of several capsid types, including AAVl, AAV2, AAV5, AAV6, AAV8, and AAV9.
  • the modified AAV vector may be based on AAV2 pseudotype with alternative muscle-tropic AAV capsids, such as AAV2/1, AAV2/6, AAV2/7, AAV2/8, AAV2/9, AAV2.5 and AAV/SASTG vectors that efficiently transduce skeletal muscle or cardiac muscle by systemic and local delivery (Seto et al. Current Gene Therapy (2012) 12:139-151).
  • AAV2 pseudotype with alternative muscle-tropic AAV capsids such as AAV2/1, AAV2/6, AAV2/7, AAV2/8, AAV2/9, AAV2.5 and AAV/SASTG vectors that efficiently transduce skeletal muscle or cardiac muscle by systemic and local delivery (Seto et al. Current Gene Therapy (2012) 12:139-151).
  • the present disclosure also provides DNA targeting systems or compositions of at least one CRISPR/Cas9-based system, as described above. These compositions may be used in genome editing, genome engineering, and correcting or reducing the effects of mutations in genes involved in genetic diseases.
  • the composition includes a CRISPR/Cas9-based system that includes a Cas9 protein or Cas9 fusion protein, as described above.
  • the CRISPR/Cas9-based system may also include at least one gRNA, as described above.
  • the present disclosure also provides multiplex CRISPR/Cas9-based systems, as described above. These compositions may be used in genome editing, genome engineering, and correcting or reducing the effects of mutations in genes involved in genetic diseases. These compositions may be used to target more than one gene.
  • the composition includes a modified lentiviral vector comprising a CRISPR/Cas9-based system that includes a Cas9 protein or Cas9 fusion protein, as described above and more than one gRNA, as described above.
  • compositions may be used to repair genetic mutations that cause disease.
  • the disclosed compositions may be used to disrupt genes such that gene disruption leads to increases in muscle regeneration or muscle strength, or decreases in muscle aging.
  • the disclosed compositions may be used to introduce therapeutic genes to be expressed systemically from skeletal muscle or cardiac muscle, such as clotting factors or monoclonal antibodies.
  • the disclosed compositions may be used to modulate mammalian gene expression.
  • the disclosed compositions may be used to transdifferentiate or induce the differentiation of a cell or correct a mutant gene in a cell. Examples of activation of genes related to cell and gene therapy, genetic reprogramming, and regenerative medicine are provided.
  • RNA-guided transcriptional activators may be used to reprogram cell lineage specification. Activation of endogenous genes encoding the key regulators of cell fate, rather than forced overexpression of these factors, may potentially lead to more rapid, efficient, stable, or specific methods for genetic reprogramming, transdifferentiation, and/or induced differentiation. 10. Methods of Genome Editing in Muscle
  • the present disclosure is directed to a method of genome editing in a skeletal muscle or cardiac muscle of a subject.
  • the method comprises administering to the skeletal muscle or cardiac muscle of the subject the composition for genome editing in skeletal muscle or cardiac muscle, as described above.
  • the genome editing may include correcting a mutant gene or inserting a transgene. Correcting the mutant gene may include deleting, rearranging, or replacing the mutant gene. Correcting the mutant gene may include nuclease-mediated NHEJ or HDR.
  • CRISPR/Cas9-based systems may be used to modulate mammalian gene expression.
  • the disclosed CRISPR/Cas9-based systems may be used to transdifferentiate or induce differentiation of a cell or correct a mutant gene in a cell.
  • RNA-guided transcriptional activators may be used to reprogram cell lineage specification. Although reprogramming was incomplete and inefficient in these experiments, there are many ways by which this method could be improved, including repeated transfections of iCas9-VP64/gRNA combinations, stable expression of these factors, and targeting multiple genes, such as Brn2 and Mytll in addition to Ascll for transdifferentiation into a neuronal phenotype.
  • RNA-guided Activation of endogenous genes encoding the key regulators of cell fate, rather than forced overexpression of these factors, may potentially lead to more rapid, efficient, stable, or specific methods for genetic reprogramming and transdifferentiation or induced differentiation of a cell.
  • Cas9 fusions to other domains, including repressive and epigenetic-modifying domains could provide a greater diversity of RNA-guided
  • transcriptional regulators to complement other RNA-based tools for mammalian cell
  • the present disclosure provides a mechanism for activating the expression of endogenous genes, such as mammalian genes, based on targeting a transcriptional activator to promoters via RNA using a CRISPR/Cas9 based system, as described above. This is
  • gRNA expression plasmids simply involves synthesizing two short custom oligonucleotides and one cloning step, it is possible to generate many new gene activators quickly and economically.
  • the gRNAs can also be transfected directly to cells following in vitro transcription. Multiple gRNAs targeted to single promoters were shown, but simultaneous targeting of multiple promoters could also be possible. Recognition of genomic target sites with RNAs, rather than proteins, may also circumvent limitations of targeting epigenetically modified sites, such as methylated DNA.
  • Cas9 fused to a transcriptional activation domain can be targeted by combinations of guide RNA molecules to induce the expression of endogenous human genes. This straightforward and versatile approach for targeted gene activation circumvents the need for engineering new proteins and allows for widespread synthetic gene regulation.
  • the method may include administering to a cell or subject a CRISPR/Cas9-based system, a polynucleotide or vector encoding said CRISPR/Cas9-based system, or DNA targeting systems or compositions of at least one CRISPR/Cas9-based system, as described above.
  • the method may include administering a CRISPR/Cas9-based system, such as administering a Cas9 fusion protein containing transcription activation domain or a nucleotide sequence encoding said Cas9 fusion protein.
  • the Cas9 fusion protein may include a transcription activation domain such as aVP16 protein or a transcription co-activator such as a p300 protein.
  • the Cas9 fusion protein may include a transcription activation domain such as aVP16 protein.
  • the transcription activation domain may contain at least 1 VP 16 protein, at least 2 VP 16 proteins, at least 3 VP 16 proteins, at least 4 VP 16 proteins (i.e., a VP64 activator domain), at least 5 VP 16 proteins, at least 6 VP 16 proteins, at least 6 VP 16 proteins, or at least 10 VP 16 proteins.
  • the Cas9 protein may be a Cas9 protein in which the nuclease activity is inactivated.
  • the Cas9 protein in the fusion protein may be iCas9 (amino acids 36-1403 of SEQ ID NO: 1), which includes the amino acid substitutions of D10A and H840A.
  • the Cas9 fusion protein may be iCas9-VP64.
  • the Cas9 fusion protein may include a transcription co-activation domain such as a p300 protein.
  • the p300 protein also known as EP300 or El A binding protein p300
  • the p300 protein plays a role in regulating cell growth and division, prompting cells to mature and assume specialized functions (differentiate) and preventing the growth of cancerous tumors.
  • the p300 protein activates transcription by connecting transcription factors with a complex of proteins that carry out transcription in the cell's nucleus.
  • the p300 interaction with transcription factors is managed by one or more of p300 domains: the nuclear receptor interaction domain (RID), the CREB and MYB interaction domain (KIX), the cysteine/histidine regions
  • the protein functions as histone acetyltransferase that regulates transcription via chromatin remodeling, and is important in the processes of cell proliferation and differentiation. It mediates cAMP-gene regulation by binding specifically to phosphorylated CREB protein.
  • the p300 protein may activate Mothers against decapentaplegic homolog 7, MAF, TSG101, Peroxisome proliferator-activated receptor alpha, NPAS2, PAX6, DDX5, MYBL2, Mothers against decapentaplegic homolog 1, Mothers against decapentaplegic homolog 2, Lymphoid enhancer-binding factor 1, SNIP1, TRERFl, STAT3, EID1, RAR-related orphan receptor alpha, ELK1, HIF1A, ING5, Peroxisome proliferator-activated receptor gamma, SS18, TCF3, Zif268, Estrogen receptor alpha, GPS2, MyoD, YY1, ING4, PROX1, CITEDl, HNF1A, MEF2C, MEF2D, MAML1, Twist transcription factor, PTMA, IRF2, DTX1, Flap structure- specific endonuclease 1, Myocyte-specific enhancer factor 2A, CDX2, BRCA1, FINRPU
  • the transcription co-activation domain may include a human p300 protein or a fragment thereof.
  • the transcription co-activation domain may include a wild-type human p300 protein or a mutant human p300 protein, or fragments thereof.
  • the transcription co-activation domain may include the core lysine-acetyltranserase domain of the human p300 protein, i.e., the p300 HAT Core (also known as "p300 WT Core"; see Fig. 58).
  • the Cas9 protein may be a Cas9 protein in which the nuclease activity is inactivated.
  • the Cas9 protein in the fusion protein may be iCas9 (amino acids 36-1403 of SEQ ID NO: 1), which includes the amino acid substitutions of D10A and H840A.
  • the Cas9 fusion protein may be iCas9-p300 WT Core. (3) gRNA
  • the method may also include administering to a cell or subject a CRISPR/Cas9-based system at least one gRNA, wherein the gRNAs target different DNA sequences.
  • the target DNA sequences may be overlapping.
  • the number of gRNA administered to the cell may be at least 1 gRNA, at least 2 different gRNAs, at least 3 different gRNAs at least 4 different gRNAs, at least 5 different gRNAs, at least 6 different gRNAs, at least 7 different gRNAs, at least 8 different gRNAs, at least 9 different gRNAs, at least 10 different gRNAs, at least 11 different gRNAs, at least 12 different gRNAs, at least 13 different gRNAs, at least 14 different gRNAs, at least 15 different gRNAs, at least 16 different gRNAs, at least 17 different gRNAs, at least 18 different gRNAs, at least 18 different gRNAs, at least 20 different gRNAs, at least 25 different
  • the number of gRNA administered to the cell may be between at least 1 gRNA to at least 50 different gRNAs, at least 1 gRNA to at least 45 different gRNAs, at least 1 gRNA to at least 40 different gRNAs, at least 1 gRNA to at least 35 different gRNAs, at least 1 gRNA to at least 30 different gRNAs, at least 1 gRNA to at least 25 different gRNAs, at least 1 gRNA to at least 20 different gRNAs, at least 1 gRNA to at least 16 different gRNAs, at least 1 gRNA to at least 12 different gRNAs, at least 1 gRNA to at least 8 different gRNAs, at least 1 gRNA to at least 4 different gRNAs, at least 4 gRNAs to at least 50 different gRNAs, at least 4 different gRNAs to at least 45 different gRNAs, at least 4 different gRNAs to at least 40 different gRNAs, at least 4 different gRNAs to at least 35
  • the gRNA may comprise a "G" at the 5' end of the complementary polynucleotide sequence.
  • the gRNA may comprise at least a 10 base pair, at least a l l base pair, at least a 12 base pair, at least a 13 base pair, at least a 14 base pair, at least a 15 base pair, at least a 16 base pair, at least a 17 base pair, at least a 18 base pair, at least a 19 base pair, at least a 20 base pair, at least a 21 base pair, at least a 22 base pair, at least a 23 base pair, at least a 24 base pair, at least a 25 base pair, at least a 30 base pair, or at least a 35 base pair complementary polynucleotide sequence of the target DNA sequence followed by NGG.
  • the gRNA may target at least one of the promoter region, the enhancer region or the transcribed region of the target gene.
  • the gRNA may include a nucleic acid sequence of at least one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, and 585-625.
  • the present disclosure provides a mechanism for repressing the expression of endogenous genes, such as mammalian genes, based on targeting genomic regulatory elements, such as distal enhancers, via RNA using a CRISPR/Cas9 based system, as described above.
  • the Cas9 fusion protein may include a transcriptional repressor, such as the KRAB repressor.
  • the Cas9 fusion protein may be dCas9-KRAB.
  • the dCas9-KRAB may additionally affect epigenetic gene regulation by recruiting heterochromatin-forming factors to the targeted locus.
  • the CRISPR/dCas9-KRAB system may be used to repress the transcription of genes, but can also be used to target genomic regulatory elements which were previously inaccessible by traditional repression methods such as RNA interference (Fig. 53B).
  • Delivering dCas9-KRAB with gRNAs targeted to a distal enhancer may disrupt expression of multiple genes regulated by the targeted enhancer (see Fig. 53C).
  • the targeted enhancer may be any enhancer for a gene such as the HS2 enhancer.
  • the present disclosure provides a mechanism for transdifferentiating or inducing differentiation of cells by activating endogenous genes via RNA using a CRISPR/Cas9-based system, as described above.
  • the CRISPR/Cas9-based system may be used to transdifferentiate cells.
  • Transdifferentiation also known as lineage reprogramming or direct conversion, is a process where cells convert from one differentiated cell type to another without undergoing an intermediate pluripotent state or progenitor cell type. It is a type of metaplasia, which includes all cell fate switches, including the interconversion of stem cells. Transdifferentiation of cells has potential uses for disease modeling, drug discovery, gene therapy and regenerative medicine.
  • Activation of endogenous genes such as BRN2, MYTIL, ASCLI, NANOG, and/or MYODl
  • CRISPR/Cas9 based system described above may lead to transdifferentiation of several cell types, such as fibroblasts, cardiomyocytes, hepatocytes, chondrocytes, mesenchymal progenitor cells, hematopoetic stem cells, or smooth muscle cells, into neuronal and myogenic phenotypes, respectively.
  • the CRISPR/Cas9-based system may be used to induce differentiation of cells, such as stem cells, cardiomyocytes, hepatocytes, chondrocytes, mesenchymal progenitor cells, hematopoetic stem cells, or smooth muscle cells.
  • stem cells such as embryonic stem cells or pluripotent stem cells, may be induced to differentiate into muscle cells or vascular endothelial cell, i.e., induce neuronal or myogenic differentiation.
  • the multiplex CRISPR/Cas9-Based System takes advantage of the simplicity and low cost of sgR A design and may be helpful in exploiting advances in high-throughput genomic research using CRISPR/Cas9 technology.
  • the single lentiviral vectors described here are useful in expressing Cas9 and numerous sgRNAs in difficult cell lines, such as primary fibroblasts described here (Fig. 47).
  • the multiplex CRISPR/Cas9-Based System may be used in the same ways as the CRISPR/Cas9-Based System described above.
  • the multiplex CRISPR/Cas9-Based System may be used to activate at least one endogenous gene in a cell.
  • the method includes contacting a cell with the modified lentiviral vector.
  • the endogenous gene may be transiently activated or stably activated.
  • the endogenous gene may be transiently repressed or stably repressed.
  • the fusion protein may be expressed at similar levels to the sgR As.
  • the fusion protein may be expressed at different levels compared to the sgRNAs.
  • the cell may be a primary human cell.
  • the multiplex CRISPR/Cas9-Based System may be used in a method of multiplex gene editing in a cell.
  • the method includes contacting a cell with the modified lentiviral vector.
  • the multiplex gene editing may include correcting a mutant gene or inserting a transgene.
  • Correcting a mutant gene may include deleting, rearranging or replacing the mutant gene.
  • Correcting the mutant gene may include nuclease-mediated non-homologous end joining or homology-directed repair.
  • the multiplex gene editing may include deleting or correcting at least one gene, wherein the gene is an endogenous normal gene or a mutant gene.
  • the multiplex gene editing may include deleting or correcting at least two genes. For example, at least two genes, at least three genes, at least four genes, at least five genes, at least six genes, at least seven genes, at least eight genes, at least nine genes, or at least ten genes may be deleted or corrected.
  • the multiplex CRISPR/Cas9-Based System may be used in a method of multiplex modulation of gene expression in a cell.
  • the method includes contacting a cell with the modified lentiviral vector.
  • the method may include modulating the gene expression levels of at least one gene.
  • the gene expression of the at least one target gene is modulated when gene expression levels of the at least one target gene are increased or decreased compared to normal gene expression levels for the at least one target gene.
  • the gene expression levels may be RNA or protein levels.
  • the present disclosure is also directed to a method of correcting a mutant gene in a subject.
  • the method comprises administering to the skeletal muscle or cardiac muscle of the subject the composition for genome editing in skeletal muscle or cardiac muscle, as described above.
  • Use of the composition to deliver the site-specific nuclease to the skeletal muscle or cardiac muscle may restore the expression of a full-functional or partially-functional protein with a repair template or donor DNA, which can replace the entire gene or the region containing the mutation.
  • the site-specific nuclease may be used to introduce site-specific double strand breaks at targeted genomic loci. Site-specific double-strand breaks are created when the site-specific nuclease binds to a target DNA sequences, thereby permitting cleavage of the target DNA. This DNA cleavage may stimulate the natural DNA-repair machinery, leading to one of two possible repair pathways: homo logy-directed repair (HDR) or the non-homologous end joining (NHEJ) pathway.
  • HDR homo logy-directed repair
  • the present disclosure is directed to genome editing with a site-specific nuclease without a repair template, which can efficiently correct the reading frame and restore the expression of a functional protein involved in a genetic disease.
  • the disclosed site-specific nucleases may involve using homology-directed repair or nuclease-mediated non-homologous end joining (NHEJ)-based correction approaches, which enable efficient correction in proliferation-limited primary cell lines that may not be amenable to homologous recombination or selection-based gene correction.
  • NHEJ non-homologous end joining
  • This strategy integrates the rapid and robust assembly of active site-specific nucleases with an efficient gene editing method for the treatment of genetic diseases caused by mutations in nonessential coding regions that cause frameshifts, premature stop codons, aberrant splice donor sites or aberrant splice acceptor sites.
  • Restoration of protein expression from an endogenous mutated gene may be through template-free NHEJ-mediated DNA repair.
  • the correction of the target gene reading frame in the genome by a transiently expressed site-specific nuclease may lead to permanently restored target gene expression by each modified cell and all of its progeny.
  • Nuclease mediated NHEJ gene correction may correct the mutated target gene and offers several potential advantages over the HDR pathway.
  • NHEJ does not require a donor template, which may cause nonspecific insertional mutagenesis.
  • NHEJ operates efficiently in all stages of the cell cycle and therefore may be effectively exploited in both cycling and post-mitotic cells, such as muscle fibers. This provides a robust, permanent gene restoration alternative to oligonucleotide-based exon skipping or pharmacologic forced read-through of stop codons and could theoretically require as few as one drug treatment.
  • NHEJ-based gene correction using a CRISPR/Cas9-based system may be combined with other existing ex vivo and in vivo platforms for cell- and gene -based therapies, in addition to the plasmid electroporation approach described here.
  • delivery of a CRISPR/Cas9- based system by mRNA-based gene transfer or as purified cell permeable proteins could enable a DNA- free genome editing approach that would circumvent any possibility of insertional mutagenesis.
  • the method as described above further includes administrating a donor template to the cell.
  • the donor template may include a nucleotide sequence encoding a full- functional protein or a partially- functional protein.
  • the donor template may include a miniaturized dystrophin construct, termed minidystrophin ("minidys”), a full- functional dystrophin construct for restoring a mutant dystrophin gene, or a fragment of the dystrophin gene that after homology-directed repair leads to restoration of the mutant dystrophin gene.
  • minidystrophin minidystrophin
  • the present disclosure is also directed to genome editing with the CRISPR/Cas9-based system to restore the expression of a full-functional or partially- functional protein with a repair template or donor DNA, which can replace the entire gene or the region containing the mutation.
  • the CRISPR/Cas9-based system may be used to introduce site-specific double strand breaks at targeted genomic loci. Site-specific double-strand breaks are created when the CRISPR/Cas9- based system binds to a target DNA sequences using the gRNA, thereby permitting cleavage of the target DNA.
  • the CRISPR/Cas9-based system has the advantage of advanced genome editing due to their high rate of successful and efficient genetic modification.
  • a CRISPR/Cas9-based system directed towards the dystrophin gene may include a gRNA having a nucleic acid sequence of any one of SEQ ID NOs: 65-115.
  • the present disclosure is directed to genome editing with CRISPR/Cas9-based system without a repair template, which can efficiently correct the reading frame and restore the expression of a functional protein involved in a genetic disease.
  • the disclosed CRISPR/Cas9- based system and methods may involve using homology-directed repair or nuclease-mediated non-homologous end joining (NHEJ)-based correction approaches, which enable efficient correction in proliferation-limited primary cell lines that may not be amenable to homologous recombination or selection-based gene correction.
  • NHEJ nuclease-mediated non-homologous end joining
  • This strategy integrates the rapid and robust assembly of active CRISPR/Cas9-based system with an efficient gene editing method for the treatment of genetic diseases caused by mutations in nonessential coding regions that cause frameshifts, premature stop codons, aberrant splice donor sites or aberrant splice acceptor sites.
  • the present disclosure provides methods of correcting a mutant gene in a cell and treating a subject suffering from a genetic disease, such as DMD.
  • the method may include administering to a cell or subject a CRISPR/Cas9-based system, a polynucleotide or vector encoding said CRISPR/Cas9-based system, or composition of said CRISPR/Cas9-based system as described above.
  • the method may include administering a CRISPR/Cas9-based system, such as administering a Cas9 protein or Cas9 fusion protein containing a second domain having nuclease activity, a nucleotide sequence encoding said Cas9 protein or Cas9 fusion protein, and/or at least one gRNA, wherein the gRNAs target different DNA sequences.
  • the target DNA sequences may be overlapping.
  • the number of gRNA administered to the cell may be at least 1 gRNA, at least 2 different gRNA, at least 3 different gRNA at least 4 different gRNA, at least 5 different gRNA, at least 6 different gRNA, at least 7 different gRNA, at least 8 different gRNA, at least 9 different gRNA, at least 10 different gRNA, at least 15 different gRNA, at least 20 different gRNA, at least 30 different gRNA, or at least 50 different gRNA, as described above.
  • the gRNA may include a nucleic acid sequence of at least one of SEQ ID NOs: 65-115. The method may involve homo logy-directed repair or non-homologous end joining.
  • the present disclosure is directed to a method of treating a subject in need thereof.
  • the method comprises administering to a tissue of a subject the composition for altering gene expression and engineering or altering genomic DNA in a cell or subject genome editing, as described above.
  • the method may comprises administering to the skeletal muscle or cardiac muscle of the subject the composition for genome editing in skeletal muscle or cardiac muscle, as described above.
  • the subject may be suffering from a skeletal muscle or cardiac muscle condition causing degeneration or weakness or a genetic disease.
  • the subject may be suffering from Duchenne muscular dystrophy, as described above. a.
  • Duchenne muscular dystrophy as described above.
  • the method may be used for correcting the dystrophin gene and recovering full-functional or partially- functional protein expression of said mutated dystrophin gene.
  • the disclosure provides a method for reducing the effects (e.g., clinical symptoms/indications) of DMD in a patient.
  • the disclosure provides a method for treating DMD in a patient.
  • the disclosure provides a method for preventing DMD in a patient.
  • the disclosure provides a method for preventing further progression of DMD in a patient.
  • compositions may comprise genetic constructs that encodes the CRISPR/Cas9-based system, as disclosed herein.
  • the genetic construct such as a plasmid, may comprise a nucleic acid that encodes the CRISPR/Cas9-based system, such as the Cas9 protein and Cas9 fusion proteins and/or at least one of the gR As.
  • the compositions, as described above may comprise genetic constructs that encodes the modified AAV vector and a nucleic acid sequence that encodes the site-specific nuclease, as disclosed herein.
  • the genetic construct, such as a plasmid may comprise a nucleic acid that encodes the site-specific nuclease.
  • compositions may comprise genetic constructs that encodes the modified lentiviral vector, as disclosed herein.
  • the genetic construct such as a plasmid, may comprise a nucleic acid that encodes the Cas9-fusion protein and at least one sgRNA.
  • the genetic construct may be present in the cell as a functioning extrachromosomal molecule.
  • the genetic construct may be a linear minichromosome including centromere, telomeres or plasmids or cosmids.
  • the genetic construct may also be part of a genome of a recombinant viral vector, including recombinant lentivirus, recombinant adenovirus, and recombinant adenovirus associated virus.
  • the genetic construct may be part of the genetic material in attenuated live microorganisms or recombinant microbial vectors which live in cells.
  • the genetic constructs may comprise regulatory elements for gene expression of the coding sequences of the nucleic acid.
  • the regulatory elements may be a promoter, an enhancer, an initiation codon, a stop codon, or a polyadenylation signal.
  • the nucleic acid sequences may make up a genetic construct that may be a vector.
  • the vector may be capable of expressing the fusion protein, such as the Cas9-fusion protein or site-specific nuclease, in the cell of a mammal.
  • the vector may be recombinant.
  • the vector may comprise heterologous nucleic acid encoding the fusion protein, such as the Cas9-fusion protein or site-specific nuclease.
  • the vector may be a plasmid.
  • the vector may be useful for transfecting cells with nucleic acid encoding the Cas9-fusion protein or site-specific nuclease, which the transformed host cell is cultured and maintained under conditions wherein expression of the Cas9-fusion protein or the site-specific nuclease system takes place.
  • Coding sequences may be optimized for stability and high levels of expression. In some instances, codons are selected to reduce secondary structure formation of the RNA such as that formed due to intramolecular bonding.
  • the vector may comprise heterologous nucleic acid encoding the CRISPR/Cas9-based system or the site-specific nuclease and may further comprise an initiation codon, which may be upstream of the CRISPR/Cas9-based system or the site-specific nuclease coding sequence, and a stop codon, which may be downstream of the CRISPR/Cas9-based system or the site-specific nuclease coding sequence.
  • the initiation and termination codon may be in frame with the CRISPR/Cas9-based system or the site-specific nuclease coding sequence.
  • the vector may also comprise a promoter that is operably linked to the CRISPR/Cas9-based system or the site- specific nuclease coding sequence.
  • the promoter operably linked to the CRISPR/Cas9-based system or the site-specific nuclease coding sequence may be a promoter from simian virus 40 (SV40), a mouse mammary tumor virus (MMTV) promoter, a human immunodeficiency virus (HIV) promoter such as the bovine immunodeficiency virus (BIV) long terminal repeat (LTR) promoter, a Moloney virus promoter, an avian leukosis virus (ALV) promoter, a
  • CMV cytomegalovirus
  • EBV Epstein Barr virus
  • RSV Rous sarcoma virus
  • the promoter may also be a promoter from a human gene such as human ubiquitin C (hUbC), human actin, human myosin, human hemoglobin, human muscle creatine, or human metalothionein.
  • the promoter may also be a tissue specific promoter, such as a muscle or skin specific promoter, natural or synthetic. Examples of such promoters are described in US Patent Application Publication No.
  • the vector may also comprise a polyadenylation signal, which may be downstream of the CRISPR/Cas9-based system or the site-specific nuclease.
  • the polyadenylation signal may be a SV40 polyadenylation signal, LTR polyadenylation signal, bovine growth hormone (bGH) polyadenylation signal, human growth hormone (hGH) polyadenylation signal, or human ⁇ - globin polyadenylation signal.
  • the SV40 polyadenylation signal may be a polyadenylation signal from a pCEP4 vector (Invitrogen, San Diego, CA).
  • the vector may also comprise an enhancer upstream of the CRISPR/Cas9-based system, i.e., the Cas9 protein or Cas9 fusion protein coding sequence or sgRNAs, or the site- specific nuclease.
  • the enhancer may be necessary for DNA expression.
  • the enhancer may be human actin, human myosin, human hemoglobin, human muscle creatine or a viral enhancer such as one from CMV, HA, RSV or EBV.
  • Polynucleotide function enhancers are described in U.S. Patent Nos. 5,593,972, 5,962,428, and WO94/016737, the contents of each are fully incorporated by reference.
  • the vector may also comprise a mammalian origin of replication in order to maintain the vector extrachromosomally and produce multiple copies of the vector in a cell.
  • the vector may also comprise a regulatory sequence, which may be well suited for gene expression in a mammalian or human cell into which the vector is administered.
  • the vector may also comprise a reporter gene, such as green fluorescent protein ("GFP") and/or a selectable marker, such as hygromycin ("Hygro").
  • the vector may be expression vectors or systems to produce protein by routine techniques and readily available starting materials including Sambrook et al., Molecular Cloning and Laboratory Manual, Second Ed., Cold Spring Harbor (1989), which is incorporated fully by reference.
  • the vector may comprise the nucleic acid sequence encoding the CRISPR/Cas9-based system, including the nucleic acid sequence encoding the Cas9 protein or Cas9 fusion protein and the nucleic acid sequence encoding the at least one gRNA
  • nucleic acid sequence comprising the nucleic acid sequence of at least one of SEQ ID NOs: 5-40, 65-144, 492-515, 540-563, and 585-625.
  • the composition may be in a pharmaceutical composition.
  • the pharmaceutical composition may comprise about 1 ng to about 10 mg of DNA encoding the CRISPR/Cas9- based system or CRISPR/Cas9-based system protein component, i.e., the Cas9 protein or Cas9 fusion protein.
  • the pharmaceutical composition may comprise about 1 ng to about 10 mg of the DNA of the modified AAV vector and nucleotide sequence encoding the site-specific nuclease.
  • the pharmaceutical composition may comprise about 1 ng to about 10 mg of the DNA of the modified lentiviral vector.
  • the pharmaceutical compositions according to the present invention are formulated according to the mode of administration to be used. In cases where
  • compositions are injectable pharmaceutical compositions, they are sterile, pyrogen free and particulate free.
  • An isotonic formulation is preferably used.
  • additives for isotonicity may include sodium chloride, dextrose, mannitol, sorbitol and lactose.
  • isotonic solutions such as phosphate buffered saline are preferred.
  • Stabilizers include gelatin and albumin.
  • a vasoconstriction agent is added to the formulation.
  • the composition may further comprise a pharmaceutically acceptable excipient.
  • the pharmaceutically acceptable excipient may be functional molecules as vehicles, adjuvants, carriers, or diluents.
  • the pharmaceutically acceptable excipient may be a transfection facilitating agent, which may include surface active agents, such as immune-stimulating complexes
  • ISCOMS Intra-Reactive Immunosarcoma
  • LPS Long Term Evolution
  • lipids liposomes
  • calcium ions calcium ions
  • viral proteins polyanions, polycations, or nanoparticles, or other known transfection facilitating agents.
  • the transfection facilitating agent is a polyanion, polycation, including poly-L- glutamate (LGS), or lipid.
  • the transfection facilitating agent is poly-L-glutamate, and more preferably, the poly-L-glutamate is present in the composition for genome editing in skeletal muscle or cardiac muscle at a concentration less than 6 mg/ml.
  • the transfection facilitating agent may also include surface active agents such as immune-stimulating complexes (ISCOMS), Freunds incomplete adjuvant, LPS analog including monophosphoryl lipid A, muramyl peptides, quinone analogs and vesicles such as squalene and squalene, and hyaluronic acid may also be used administered in conjunction with the genetic construct.
  • ISCOMS immune-stimulating complexes
  • LPS analog including monophosphoryl lipid A
  • muramyl peptides muramyl peptides
  • quinone analogs and vesicles such as squalene
  • the DNA vector encoding the composition may also include a transfection facilitating agent such as lipids, liposomes, including lecithin liposomes or other liposomes known in the art, as a DNA-liposome mixture (see for example W09324640), calcium ions, viral proteins, polyanions, polycations, or nanoparticles, or other known transfection facilitating agents.
  • the transfection facilitating agent is a polyanion, polycation, including poly-L-glutamate (LGS), or lipid. 17.
  • compositions may be the transfection or electroporation of the composition as a nucleic acid molecule that is expressed in the cell and delivered to the surface of the cell.
  • the nucleic acid molecules may be electroporated using BioRad Gene Pulser Xcell or Amaxa Nucleofector lib devices.
  • Several different buffers may be used, including BioRad electroporation solution, Sigma phosphate-buffered saline product #D8537 (PBS), Invitrogen OptiMEM I (OM), or Amaxa Nucleofector solution V (N.V.).
  • Transfections may include a transfection reagent, such as Lipofectamine 2000.
  • the transfected cells Upon delivery of the composition to the tissue, and thereupon the vector into the cells of the mammal, the transfected cells will express the fusion protein, such as a CRISPR/Cas9- based system and/or a site-specific nuclease.
  • the composition may be administered to a mammal to alter gene expression or to re-engineer or alter the genome.
  • the composition may be administered to a mammal to correct the dystrophin gene in a mammal.
  • the mammal may be human, non-human primate, cow, pig, sheep, goat, antelope, bison, water buffalo, bovids, deer, hedgehogs, elephants, llama, alpaca, mice, rats, or chicken, and preferably human, cow, pig, or chicken.
  • the vector encoding a CRISPR/Cas9-based system protein component i.e., the Cas9 protein or Cas9 fusion protein
  • a CRISPR/Cas9-based system protein component i.e., the Cas9 protein or Cas9 fusion protein
  • the recombinant vector may be delivered by any viral mode.
  • the viral mode may be recombinant lentivirus, recombinant adenovirus, and/or recombinant adeno-associated virus.
  • the nucleotide encoding a CRISPR/Cas9-based system protein component may be introduced into a cell to genetically correct the target gene or alter gene expression of a gene, such as activate or repress endogenous genes.
  • a nucleotide encoding a CRISPR/Cas9-based system protein component, i.e., the Cas9 protein or Cas9 fusion protein, directed towards a mutant dystrophin gene by the gRNA may be introduced into a myoblast cell from a DMD patient.
  • the genetically corrected fibroblast cell may be treated with MyoD to induce differentiation into myoblasts, which may be implanted into subjects, such as the damaged muscles of a subject to verify that the corrected dystrophin protein was functional and/or to treat the subject.
  • the modified cells may also be stem cells, such as induced pluripotent stem cells, bone marrow-derived progenitors, skeletal muscle progenitors, human skeletal myoblasts from DMD patients, CD 133+ cells, mesoangioblasts, and MyoD- or Pax7- transduced cells, or other myogenic progenitor cells.
  • the CRISPR/Cas9-based system may cause neuronal or myogenic differentiation of an induced pluripotent stem cell.
  • compositions may be administered to a subject by different routes including orally, parenterally, sublingually, transdermally, rectally, transmucosally, topically, via inhalation, via buccal administration, intrapleurally, intravenous, intraarterial, intraperitoneal, subcutaneous, intramuscular, intranasal intrathecal, and intraarticular or combinations thereof.
  • the composition may be administered as a suitably acceptable formulation in accordance with normal veterinary practice. The veterinarian may readily determine the dosing regimen and route of administration that is most appropriate for a particular animal.
  • the compositions may be administered by traditional syringes, needleless injection devices,
  • microprojectile bombardment gone guns or other physical methods such as electroporation (“EP), “hydrodynamic method”, or ultrasound.
  • EP electroporation
  • hydrodynamic method or ultrasound.
  • the composition may be delivered to the mammal by several technologies including DNA injection (also referred to as DNA vaccination) with and without in vivo electroporation, liposome mediated, nanoparticle facilitated, recombinant vectors such as recombinant lentivirus, recombinant adenovirus, and recombinant adenovirus associated virus.
  • DNA injection also referred to as DNA vaccination
  • liposome mediated such as recombinant lentivirus, recombinant adenovirus, and recombinant adenovirus associated virus.
  • the composition may be injected into the skeletal muscle or cardiac muscle.
  • the composition may be injected into the tibialis anterior muscle.
  • Cell types may be fibroblasts, pluripotent stem cells, cardiomyocytes, hepatocytes, chondrocytes, mesenchymal progenitor cells, hematopoetic stem cells, smooth muscle cells, or K562 human erythroid leukemia cell line.
  • Cell types currently under investigation for cell-based therapies of DMD include immortalized myoblast cells, such as wild-type and DMD patient derived lines, for example ⁇ 48- 50 DMD, DMD 8036 (del48-50), C25C14 and DMD-7796 cell lines, primal DMD dermal fibroblasts, induced pluripotent stem cells, bone marrow-derived progenitors, skeletal muscle progenitors, human skeletal myoblasts from DMD patients, CD 133+ cells, mesoangioblasts, cardiomyocytes, hepatocytes, chondrocytes, mesenchymal progenitor cells, hematopoetic stem cells, smooth muscle cells, and MyoD- or Pax7-transduced cells, or other myogenic progenitor cells.
  • immortalized myoblast cells such as wild-type and DMD patient derived lines, for example ⁇ 48- 50 DMD, DMD 8036 (del48-50), C25C14 and DMD-7796 cell lines, prima
  • Immortalization of human myogenic cells may be used for clonal derivation of genetically corrected myogenic cells.
  • Cells may be modified ex vivo to isolate and expand clonal populations of immortalized DMD myoblasts that contain a genetically corrected dystrophin gene and are free of other nuclease-introduced mutations in protein coding regions of the genome.
  • transient in vivo delivery of nucleases by non-viral or non-integrating viral gene transfer, or by direct delivery of purified proteins and gR As containing cell- penetrating motifs may enable highly specific correction in situ with minimal or no risk of exogenous DNA integration.
  • kits which may be used to edit a genome in skeletal muscle or cardiac muscle, such as correcting a mutant gene.
  • the kit comprises a composition for genome editing in skeletal muscle or cardiac muscle, as described above, and instructions for using said composition.
  • Instructions included in kits may be affixed to packaging material or may be included as a package insert. While the instructions are typically written or printed materials they are not limited to such. Any medium capable of storing such instructions and
  • Such media include, but are not limited to, electronic storage media ⁇ e.g., magnetic discs, tapes, cartridges, chips), optical media ⁇ e.g., CD ROM), and the like.
  • instructions may include the address of an internet site that provides the instructions.
  • the composition for genome editing in skeletal muscle or cardiac muscle may include a modified AAV vector and a nucleotide sequence encoding a site-specific nuclease, as described above.
  • the site-specific nuclease may include a ZFN, a TALEN, or CRISPR/Cas9-based system, as described above, that specifically binds and cleaves a mutated gene.
  • the site-specific nuclease, as described above may be included in the kit to specifically bind and target a particular region in the mutated gene.
  • the site-specific nuclease may be specific for a mutated dystrophin gene, as described above.
  • the kit may further include donor DNA, a gRNA, or a transgene, as described above.
  • kits which may be used to correct a mutated gene.
  • the kit comprises at least one component for correcting a mutated gene and instructions for using the CRISPR/Cas9-based system.
  • Instructions included in kits may be affixed to packaging material or may be included as a package insert. While the instructions are typically written or printed materials they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this disclosure. Such media include, but are not limited to, electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like.
  • the term "instructions" may include the address of an internet site that provides the instructions.
  • At least one component may include at least one CRISPR/Cas9-based system, as described above, which specifically targets a gene.
  • the kit may include a Cas9 protein or Cas9 fusion protein, a nucleotide sequence encoding said Cas9 protein or Cas9 fusion protein, and/or at least one gRNA.
  • the CRISPR/Cas9-based system, as described above, may be included in the kit to specifically bind and target a particular target region upstream, within or downstream of the coding region of the target gene.
  • a CRISPR/Cas9-based system may be specific for a promoter region of a target gene or a CRISPR/Cas9-based system may be specific for a mutated gene, for example a mutated dystrophin gene, as described above.
  • the kit may include donor DNA, as described above.
  • HEK293T cells were obtained from the American Tissue Collection Center (ATCC) through the Duke University Cancer Center Facilities and were maintained in DMEM supplemented with 10% fetal bovine serum and 1%
  • HEK293T cells were transfected with
  • Lipofectamine 2000 (Invitrogen) according to manufacturer's instructions. Transfection efficiencies were routinely higher than 80% as determined by fluorescence microscopy following delivery of a control eGFP expression plasmid. Cas9 expression plasmid was transfected at a mass ratio of 3 : 1 to either the individual gRNA expression plasmids or the identical amount of gR A expression plasmid consisting of a mixture of equal amounts of the four gR As.
  • PMEF-HL Primary mouse embryonic fibroblasts (PMEF-HL, Millipore, Billerica, MA) were seeded (75,000 per well) in 24-well TCPS plates (BD, Franklin Lakes, NJ) and maintained at 37°C and 5% C0 2 in complete MEF medium consisting of high glucose DMEM supplemented with 10% Premium Select FBS (Atlanta Biologicals, LawrenceviUe, GA), 25 ⁇ g mL "1 gentamicin (Invitrogen), IX GlutaMAX, non-essential amino acids, sodium pyruvate, and ⁇ - mercaptoethanol (Invitrogen).
  • MEF transfections were performed with a single 1 ⁇ g cm "2 dose of total plasmid DNA, delivered as cationic nanocomplexes following electrostatic condensation with poly(CBA-ABOL) in serum- and antibiotic-free OptiMEM, as described previously (Adler et al. Molecular therapy. Nucleic acids 1, e32 (2012)). OptiMEM was replaced with complete MEF medium 4 hrs after transfection. 48 hrs after transfection, MEFs were processed for qRT- PCR, or the complete MEF medium was replaced with N3 neural induction medium containing: DMEM/F-12 (Invitrogen), IX N-2 Supplement (Invitrogen), 10 ng mL "1 human bFGF2
  • a GFP reporter vector (pmax-GFP, 3486 bp, Amaxa, Cologne, Germany) was used to optimize transfection conditions.
  • Cas9 expression plasmid was transfected at a mass ratio of 3 : 1 or 1 : 1 to an equal mixture of four gR A expression plasmids.
  • Plasmids The plasmids encoding wild-type and H840A Cas9 were obtained from Addgene (Plasmid #39312 and Plasmid #39316; Jinek, et al. Science 337, 816-821 (2012)). H840A Cas9 was cloned into the vector pcDNA3.1 in frame with a FLAG epitope tag and a nuclear localization sequence (NLS) at the N-terminus with a primer pair that introduced the DIOA mutation. The VP64 domain, an NLS, and an HA epitope tag were cloned in frame with the Cas9 ORF at the C-terminus (Fig. la, Fig. 9a).
  • the tracrRNA and crRNA expression cassettes (Cong et al. Science 339, 819-823 (2013)) were ordered as gBlocks (Integrated DNA Technologies (IDT)) and cloned into a pZDonor plasmid (Sigma) with Kpnl and SacII sites.
  • a chimeric guide RNA expression cassette (Mali et al. Science 339, 823-826 (2013)) was also ordered as gBlocks with modifications to include a Bbsl restriction site to facilitate rapid cloning of new guide RNA spacer sequences (Fig. 9b).
  • the oligonucleotides containing the target sequences were obtained from IDT, hybridized, phosphorylated, and cloned in the appropriate plasmids using Bbsl sites.
  • the target sequences are provided in Table 2.
  • Membranes labeled with primary antibodies were incubated with anti-rabbit HRP-conjugated antibody (Sigma- Aldrich) diluted 1 :5000 for 30 min, anti-goat (1 :3000) or anti-mouse (1 :5000) and washed with TBS-T for 30 min. Membranes were visualized using the Immun-Star WesternCTM
  • ELISA Serum-free culture media (OPTI-MEM) was collected and frozen at -80°C. Human IL-lra secretion into culture media was quantified via enzyme-linked immunosorbent assay (ELISA), according to the manufacturer's protocols (R&D Systems, Cat. No. DY280). The standard curve was prepared by diluting recombinant human IL-lra in OPTI-MEM and the IL-lra in culture media was measured undiluted. The samples were concentrated about 8 fold via centrifugation through 3 kDa MWCO filters for 20 min (Amicon Ultra, Cat # UFC500396). Reported values were corrected by the concentration factor for each sample.
  • RNA-Seq RNA seq libraries were constructed. Briefly, first strand cDNA was synthesized from oligo dT Dynabead® (Invitrogen) captured mRNA using Superscript®
  • Second strand cDNA was synthesized using DNA Polymerase I (New England Biolabs). cDNA was purified using Agencourt AMPure XP beads (Beckman Coulter) and Nextera transposase (Illumina; 5 min at 55 °C) was used to
  • the cells were then washed three times with PBS, incubated for 1 hr at room temperature in blocking buffer with Alexa Fluor 488 goat anti-mouse IgG and Alexa Fluor 594 goat anti-rabbit IgG (Invitrogen, 1 :200), and washed three times with PBS.
  • Stained MEFs were then scanned with a Nikon Eclipse TE2000-U inverted fluorescence microscope with a ProScanll motorized stage (Prior Scientific, Rockland, MA) to produce large mosaic images of each complete culture area.
  • These mosaics were processed with a FIJI macro to automatically and uniformly threshold each image according to local contrast, exclude small debris, and to count the number of Tuj l + cells in each well.
  • the CRISPR system recognizes its target through base pairing of a 20 bp sequence in the gRNA to a complementary DNA target, which is followed by the NGG protospacer-adjacent motif (PAM) sequence, where N is any base pair.
  • PAM protospacer-adjacent motif
  • the four target site sequences were introduced into crRNA and gRNA expression plasmids 17 and co-transfected with the iCas9-VP64 expression plasmid into HEK293T cells. Although substantial induction of IL1RN expression was observed by qRT-PCR in samples treated with the combination of crRNAs, much higher levels were achieved with the combination of gRNAs (Fig. lc). No changes to gene expression were observed in cells treated with gRNAs and an expression plasmid for iCas9 without VP64, demonstrating the critical role of the activation domain in modulating gene expression (Fig. lc).
  • Nuclease activity at these target sites was confirmed to have been abrogated in the iCas9-VP64 system by performing the Surveyor assay to detect DNA repair events in samples treated with iCas9-VP64 and wild-type Cas9 (Fig. 5).
  • Fig. Id High levels of IL1RN expression were observed only when the gRNA combinations were co-transfected with iCas9-VP64 (Fig. Id), as seen with other classes of engineered transcription factors.
  • IL-1 receptor antagonist IL-1 receptor antagonist
  • gRNAs were designed to target each of the promoters of eight other genes relevant to medicine and biotechnology, including ASCL1, NANOG, HBG1/2, MYOD1, VEGFA, TERT, IL1B, and IL1R2 (Fig. 4, Table 2). Forced expression of ASCL1 and MYOD1 leads to transdifferentiation of several cell types into neuronal and myogenic phenotypes, respectively.
  • NANOG is a marker of pluripotency and that is also used in genetic reprogramming strategies.
  • Activation of the homologs HBG1 and HBG2, which encode ⁇ -globin during fetal development, can be used as a therapeutic strategy to compensate for ⁇ -globin mutations in sickle cell disease.
  • Up-regulation of VEGFA by synthetic transcription factors has been explored as a strategy to enhance tissue regeneration and wound healing.
  • the forced expression of telomerase, encoded by the TERT gene can be used to immortalize cell lines.
  • IL1B encodes the IL- ⁇ cytokine that mediates inflammation and autoimmunity.
  • IL- ⁇ signaling can be blocked by expression of IL-lra or the decoy receptor encoded by IL1R2.
  • RNA-seq was performed on cells transfected with iCas9- VP64 and the four gRNAs targeting HBG1, three of which also target HBG2.
  • iCas9-VP64 As preliminary evidence that the activation of gene targets by iCas9-VP64 can lead to secondary changes in gene networks and cell phenotypes, expression plasmids for iCas9-VP64 and the four gRNAs targeting ASCLI were co-transfected into murine embryonic fibroblasts (MEFs) (Fig. 8). Forced expression of Ascll in MEFs has been shown to partially activate the neuronal gene network, including the downstream target Tuj 1. Because the gRNA target sites are conserved in the human and mouse ASCLI promoters (Fig. 8a), activation of ASCLI expression was also observed in MEFs treated with plasmids encoding iCas9-VP64 and the four gRNAs (Fig. 8b).
  • IL1RN and HBG1/2 were chosen for this specificity analysis as the gene products, IL-lra and ⁇ -globin, may not generate secondary effects on gene expression in HEK293T cells. Exploiting the synergistic activity of multiple weak transcriptional activators, in contrast to using a single strong activator, may increase specific gene regulation since it is unlikely that multiple adjacent off- target sites would exist at another locus. Interestingly, the IL32 gene was moderately
  • Plasmid constructs The expression cassettes for the S. pyogenes sgRNA and human codon optimized Cas9 (hCas9) nuclease were used, as previously described (Perez-Pinera et al., Nat Methods 10:973-976 (2013)).
  • a GeneBlock IDT was synthesized containing a portion of the 3' end of the Cas9 coding sequence fused to a T2A skipping peptide immediately upstream of a multiple cloning site and subsequently cloned into the hCas9 expression vector.
  • An eGFP reporter gene was then cloned into the T2A vector to allow co-translation of Cas9 and eGFP proteins from the same expression vector (hCas9-T2A-GFP, SEQ ID NO: 116).
  • HEK293T cells were obtained from the American Tissue Collection Center (ATCC) through the Duke Cell Culture Facility and were maintained in DMEM supplemented with 10% bovine calf serum and 1% penicillin/streptomycin.
  • Immortalized myoblasts (Mamchaoui, K. et al. Skelet Muscle 1, 1-11 (2011)) (one from a wild- type donor, and two ⁇ 48-50 DMD patient derived lines) were maintained in skeletal muscle media (PromoCell) supplemented with 20%> bovine calf serum (Sigma), 50 ⁇ g/ml fetuin, 10 ng/ml human epidermal growth factor (Sigma), 1 ng/ml human basic fibroblast growth factor (Sigma), 10 ⁇ g/ml human insulin (Sigma), 1% GlutaMAX (Invitrogen), and 1%
  • Primary DMD dermal fibroblasts were obtained from the Coriell Cell repository (GM05162A, ⁇ 46-50) and maintained in DMEM supplemented with 10% fetal bovine serum, 1 ng/mL human basic fibroblast growth factor, and 1%
  • HEK293T cells were transfected with Lipofectamine 2000 (Invitrogen) with 400 ng of each expression vector according to the manufacturer's protocol in 24 well plates.
  • Immortalized myoblasts and primary fibroblasts were transfected with 5 ⁇ g of each expression vector by electroporation using the Gene Pulser XCell (BioRad) with PBS as an electroporation buffer using optimized conditions for each line (Fig. 1) (Ousterout et al. Mol Ther 21 : 1718-1726 (2013)).
  • Transfection efficiencies were measured by delivering an eGFP expression plasmid (pmaxGFP, Clontech) and using flow cytometry.
  • the indicated mass of electroporated plasmid corresponds to the amount used for each CRISPR/Cas9-based system.
  • CRISPR/Cas9-based system-induced lesions at the endogenous target site were quantified using the Surveyor nuclease assay (Guschin, D.Y. et al. Meth Mol Biol 649, 247-256 (2010)), which can detect mutations characteristic of nuclease-mediated NHEJ. After transfection, cells were incubated for 3 or 10 days at 37°C and genomic DNA was extracted using the DNeasy Blood and Tissue kit (QIAGEN).
  • the target locus was amplified by 35 cycles of PCR with the AccuPrime High Fidelity PCR kit (Invitrogen) using primers specific to each locus (see Table 4), such as 5'- GAGTTTGGCTCAAATTGTTACTCTT-3 ' (SEQ ID NO: 60) and 5'- GGGAAATGGTCTAGGAGAGTAAAGT-3 ' (SEQ ID NO: 61).
  • Table 4 Summary of top 10 off target sites predicted in silico and activity at each site as detected by the Surveyor assay in HEK293T cells transfected with Cas9 and the indicated sgRNA expression cassettes, n.d.: not detected.
  • PCR products were randomly melted and reannealed in a thermal cycler with the program: 95°C for 240 s, followed by 85°C for 60 s, 75°C for 60 s, 65°C for 60 s, 55°C for 60 s, 45°C for 60 s, 35°C for 60 s, and 25°C for 60 s with a -0.3°C/s rate between steps.
  • 8 ⁇ , of PCR product was mixed with 1 ⁇ , of Surveyor Nuclease S and 1 ⁇ ⁇ of Enhancer S (Transgenomic) and incubated at 42°C for 1 hr.
  • PCR-based assay to detect genomic deletions The exon 51 or exon 45-55 loci were amplified from genomic DNA by PCR (Invitrogen AccuPrime High Fidelity PCR kit) using primers flanking each locus.
  • the flanking primers were CelI-CRl/2-F and CelI-CR5-R for exon 51 or CelI-CR6-F and CelI-CR36-R for exon 45-55 analysis (Table 4).
  • PCR products were separated on TAE-agarose gels and stained with ethidium bromide for analysis.
  • PCR-based detection of translocations Loci with predicted possible translocations were amplified by a two-step nested PCR (Invitrogen AccuPrime High Fidelity PCR kit for each step) of genomic DNA from cells transfected with Cas9 alone (control) or Cas9 with sgRNA.
  • translocations that may occur at each on-target and off-target sgRNA target site were amplified by 35 cycles of PCR using combinations of Surveyor primers for each locus that were modified to include restriction sites to facilitate cloning and sequencing analysis (Table 4).
  • PCR amplicons corresponding to the expected length of predicted translocations and only present in cells treated with sgRNA were purified (QIAGEN Gel Extraction kit) and analyzed by Sanger sequencing.
  • RNA analysis Immortalized myoblasts were differentiated into myofibers by replacing the growth medium with DMEM supplemented with 1% insulin-transferrin-selenium (Invitrogen #51500056) and 1% penicillin/streptomycin (Invitrogen #15140) for 5 days before the cells were trypsinized and collected. Total RNA was isolated from these cells using the RNeasy Plus Mini Kit (QIAGEN) according to the manufacturer's instructions. RNA was reverse transcribed to cDNA using the VILO cDNA synthesis kit (Life Technologies #11754) and 1.5 micrograms of RNA for 2 hrs at 42°C according to the manufacturer's instructions.
  • the target loci were amplified by 35 cycles of PCR with the AccuPrime High Fidelity PCR kit (Invitrogen) using primers annealing to exons 44 and 52 to detect exon 51 deletion by CRl/5 or CR2/5 or primers annealing to exons 44 and 60 to detect exon 45-55 deletion by CR6/36 (Table 4).
  • PCR products were run on TAE-agarose gels and stained with ethidium bromide for analysis. The resolved PCR bands were cloned and analyzed by Sanger sequencing to verify the expected exon junctions.
  • Table 5 lists the sequences of primers used in Example 4.
  • Blots were probed with the following primary antibodies: MANDYS8 to detect dystrophin (1 :100, Sigma D8168) and rabbit anti-GAPDH (1 :5000, Cell Signaling 2118S). Blots were then incubated with mouse or rabbit horseradish peroxidase- conjugated secondary antibodies (Santa Cruz) and visualized using the ChemiDoc
  • mice Transplantation into immunodeficient mice. All animal experiments were conducted under protocols approved by the Duke Institutional Animal Care & Use Committee. Cells were trypsinized, collected and washed in IX Hank's Balanced Salt Solution (HBSS, Sigma). Two million cells were pelleted and resuspended in five IX HBSS (Sigma) supplemented with cardiotoxin (Sigma #C9759) immediately prior to injection. These cells were transplanted into the hind limb tibialis anterior (TA) muscle of NOD.SCID.gamma (NSG) mice (Duke CCIF Breeding Core) by intramuscular injection. Four weeks after injection, mice were euthanized and the TA muscles were harvested.
  • HBSS IX Hank's Balanced Salt Solution
  • Cryosections were incubated overnight at 4°C with the following primary antibodies that are specific to human epitopes only: anti-spectrin (1 :20, Leica NCL- SPEC1) or anti-dystrophin (1 :2, Leica NCL-DYS3). After primary staining, spectrin or dystrophin expression was detected using a tyramide-based immunofluorescence signal amplification detection kit (Life Technologies, TSA Kit #22, catalog #T-20932,). Briefly, cryosections were incubated with 1 :200 goat anti-mouse biotin-XX secondary (Life Technologies, TSA Kit #22, catalog #T-20932,). Briefly, cryosections were incubated with 1 :200 goat anti-mouse biotin-XX secondary (Life Technologies, TSA Kit #22, catalog #T-20932,). Briefly, cryosections were incubated with 1 :200 goat anti-mouse biotin-XX secondary (Life Technologies, TSA Kit #22, catalog #T-20
  • Cytotoxicity assay To quantitatively assess potential sgRNA or SpCas9 nuclease- associated cytotoxicity, HEK293T cells were transfected with 10 ng of a GFP reporter and 100 ng SpCas9 expression vector and 100 ng sgRNA expression vector using Lipofectamine 2000 according to the manufacturer's instructions (Invitrogen). The percentage of GFP positive cells was assessed at 2 and 5 days by flow cytometry. The survival rate was calculated as the decrease in GFP positive cells from days 2 to 5 and normalized to cells transfected with an empty nuclease expression vector as described (Cornu et al, Meth Mol Biol 649:237-245 (2010)).
  • the CRISPR/Cas9-based system was designed to target the dystrophin gene.
  • Various gRNAs were chosen to target different regions of the human and mouse dystrophin gene based on NNNNN NNNNN NNNNN NNNNN NGG and GNNNN NNNNN NNNNN NNNNN NGG (see Tables 6, 7 and 8).
  • NNNNGANN Target 3' splice donor of exon NmCas9 NNNNGANN
  • NNNNGANN Target 3' splice donor of exon NmCas9 NNNNGANN
  • NNNNGANN Target 3' splice donor of exon NmCas9 NNNNGANN
  • NNNNGANN Frameshift in exon 51 NmCas9 NNNNGANN
  • NNNNGANN Delete exon 51 (bind as close NmCas9 (NNNNGANN
  • NNNNGANN Delete exon 51 (bind as close NmCas9 (NNNNGANN
  • NNNNGANN Delete exons 45-55, bind as NmCas9 (NNNNGANN
  • N NGTTN Delete exons 45-55 bind as NmCas9 (NNNNGTTN
  • N NGN T Delete exons 45-55, bind as NmCas9 (NNNNGNNT
  • NNNNGANN Delete exons 45-55, bind as NmCas9 (NNNNGANN
  • the CRISPR/Cas9-based system was used in DMD 8036 (del48-50) cells to determine if the system could repair a mutant dystrophin gene.
  • 5 ⁇ g of Cas9 was co-transfected into DMD 8036 (del48-50) cells with either 7.5 ⁇ g of empty vector or gRNA.
  • 7.5 ⁇ g of CRl (“DCR1")
  • 7.5 ⁇ g of CR5 (“DCR5"
  • DCR3 15 ⁇ g of CR3
  • DCR1+DCR5 7.5 ⁇ g of a combination of CRl and CR5
  • Fig. 13 This locus was amplified by PCR using primers flanking the region containing the genomic targets for CRl and CR5 (the forward primer: 5'-gagaggttatgtggctttacca (SEQ ID NO:457), the reverse primer: 5'-ctgcgtagtgccaaaacaaa (SEQ ID NO:458)), resulting in a 1447 bp band for the wild-type locus or an expected size of approximately 630 bp for the deleted locus. After 7 days of differentiation, western blot of the treated cells shows expression of dystrophin protein (see Fig. 14).
  • sgRNAs targeted to the hotspot mutation region between exons 45-55 were created (Fig. 16).
  • S. pyogenes system that utilizes a human-codon optimized SpCas9 nuclease and a chimeric single-guide RNA (sgRNA) expression vector to guide efficient site-specific gene editing was used. Similar to Example 4 targeting exon 51 with TALENs, protospacers were selected to target the 5' and 3' ends of exons 45 through 55 which meet the 5'- NRG-3' PAM requirement of SpCas9.
  • CR3 was designed to correct dystrophin mutations or deletions surrounding exon 51 by introducing small insertions or deletions in the 5' end of exon 51 to restore the downstream dystrophin reading frame (Fig. 16B).
  • sgRNAs were designed to employ the multiplex capability of the
  • sgRNAs were targeted to the intronic regions surrounding exon 51 (Fig. 16C) or exons 45-55 (Fig. 16D). These sgRNAs were intentionally targeted to sites nearest to the downstream or upstream exon intended to be included in the resulting transcript to minimize the likelihood that the background patient deletion would include the intronic sgRNA target sites.
  • Gene editing frequency in the human HEK293T cell line was assessed to rapidly determine different sgRNA targeting efficiencies.
  • HEK293Ts were transfected with constructs encoding human codon-optimized SpCas9 and the indicated sgRNA.
  • Each sgRNA was designed to modify the dystrophin gene as indicated.
  • the frequency of gene modification at day 3 or day 10 post-transfection was determined by the Surveyor assay.
  • the ratio of measured Surveyor signal at day 3 and day 10 was calculated to quantify the stability of gene editing frequencies for each sgRNA in human cells.
  • sgRNAs were selected to correct specific mutations in DMD patient myoblast cell lines. After transfection into DMD myoblasts, unexpectedly low or undetectable gene modification activity was observed as measured by the Surveyor assay (Fig. 19C, bulk population). Flow cytometry was used to select for trans fected cells co-expressing GFP through a 2A ribosomal skipping peptide linked to the SpCas9 protein (Fig. 19A). The addition of this fluorescent reporter to the SpCas9 expression vector did not seem to significantly impact gene editing activity in HEK293T cells (Fig. 19B).
  • Small insertions and deletions created by NHEJ DNA repair may be used to create targeted frameshifts to correct aberrant reading frames.
  • a sgRNA, CR3 was designed to restore the dystrophin reading frame by introducing small insertions and deletions within exon 51 (Figs. 16B, 20A).
  • the types of insertions and deletions generated by CRISPR/Cas9 at this locus were assessed by Sanger sequencing of alleles from the genomic DNA of HEK293T cells co- transfected with expression plasmids for SpCas9 and the CR3 sgRNA (Fig. 20B). Notably, the insertions and deletions resulted in conversion to all three reading frames (Figs. 20B, 20C).
  • expression plasmids for SpCas9 and the CR3 sgRNA were electroporated into a DMD myoblast line with a deletion of exons 48-50 that is correctable by creating frameshifts in exon 51.
  • the treated cells were sorted, verified to have gene modification activity by the Surveyor assay (CR3, Fig. 19C sorted population), and differentiated into myotubes to test for restored dystrophin expression. Expression of dystrophin protein was observed concomitant with the detectable nuclease activity (Fig. 20D). The S.
  • pyogenes CRISPR/Cas9 system presents a powerful method to quickly generate targeted frameshifts to address a variety of patient mutations and restore expression of the human dystrophin gene.
  • the multiplexing capability of the CRISPR/Cas9 system presents a novel method to efficiently generate genomic deletions of specific exons for targeted gene correction.
  • DMD patient myoblasts with background deletions correctable by exon 51 skipping were treated with two combinations of sgR As flanking exon 51 (CR1/CR5 or CR2/CR5) and sorted to enrich for gene-edited cells as in Fig. 19.
  • the expected genomic deletions were only present when both sgRNAs were electroporated into the cells with SpCas9 (Fig. 21 A).
  • Sanger sequencing confirmed the expected junction of the distal chromosomal segments (Fig. 21B) for both deletions.
  • this deletion was detected by PCR of the genomic DNA from SpCas9/sgRNA-treated DMD patient cells harboring a background deletion of exons 48-50 of unknown length (Fig. 22A).
  • Sanger sequencing of this deletion band from the genomic DNA of treated DMD cells revealed the expected junctions of intron 44 and intron 55 immediately adjacent to the sgRNA target sites (Fig. 22B).
  • the expected deletion of exons 45-55 was detected in the dystrophin mRNA transcript and verified to be a fusion of exons 44 and 56 by Sanger sequencing (Fig. 22C).
  • a promising method for DMD therapy is to correct a population of autologous patient muscle progenitor cells that can be engrafted into the patient's skeletal muscle tissue to rescue dystrophin expression.
  • a population of DMD myoblasts that were treated with sgRNAs CR1 and CR5, which flank exon 51, was transplanted and sorted for expression of GFP as before (Fig. 19, Fig. 23).
  • muscle fibers positive for human spectrin which is expressed by both corrected and uncorrected cells, were detected in cryosections of injected muscle tissue (Fig. 24).
  • HEK293T cell line that also shows high levels of off-target activity (Fig. 26C and Fig. 28A, 28B).
  • Sanger sequencing of the PCR amplicons confirmed the identity of the predicted translocation event for each primer pair (Figs. 29-30).
  • a subset of the translocations detected in the HEK293T cells were also detectable by nested PCR in the sorted hDMD myoblasts, although the signal was considerably weaker and the sequence identity was not confirmed due to low yield of product (Fig. 26D and Figs. 28A, 28C). Translocations were not detected using this assay in HEK293T or sorted hDMD cells treated with CR6 or CR6/CR36, respectively, (Fig.
  • Genome editing is a powerful tool for correcting genetic disease and the recent development of the CRISPR/Cas9 system is dramatically accelerating progress in this area.
  • the correction of DMD the most common genetic disease that also currently has no approved therapeutic options, was demonstrated.
  • Many gene- and cell-based therapies for DMD are in preclinical development and clinical trials, and genome editing methods are compatible with many of these approaches.
  • genome editing may be combined with patient-specific cell-based therapies for DMD.
  • the CRISPR/Cas9 system may function in human pluripotent stem cells and other human cell lines, as well as human skeletal myoblasts, as shown.
  • an enriched pool of gene-corrected cells demonstrated expression of human dystrophin in vivo following engraftment into immunodeficient mice.
  • CRISPR/Cas9 gene editing did not have significant toxic effects in human myoblasts as observed by stable gene editing frequencies and minimal cytotoxicity of several sgRNAs.
  • gene editing activity was confirmed at three out of 50 predicted off-target sites across five sgRNAs and
  • CRISPR/Cas9-induced chromosomal translocations between on-target and off-target sites were detectable.
  • the CRISPR/Cas9 technology is an efficient and versatile method for correcting a significant fraction of dystrophin mutations and can serve as a general platform for treating genetic disease.
  • sgRNA and Cas9 mRNA in contrast to the plasmid-based delivery method used here, may be used to increase specificity and safety by reducing the duration of Cas9 expression and eliminating the possibility of random plasmid integration.
  • delivery of the CRISPR/Cas9 system directly to skeletal and/or cardiac muscle by viral, plasmid, or R A delivery vectors may be used for in vivo genome editing and translation of this approach.
  • the large size of S. pyogenes Cas9 gene ( ⁇ 4.2 kilobases) presents a challenge to its use in size-restricted adeno-associated viral vectors.
  • Cas9 genes from other species such as N. meningitidis and S. thermophilus, are short enough to efficiently package both Cas9 and sgRNA expression cassettes into single AAV vectors for in vivo gene editing applications.
  • the CRISPR/Cas9 system enabled efficient modification of nearly 90% of tested targets, consistent with other reports of robust activity of this system at diverse loci.
  • the robustness and versatility of this technology is a significant advancement towards at-will creation of patient-specific gene editing.
  • Low levels of dystrophin, including as little as 4% of wild-type expression, may be sufficient to improve survival, motor function, and cardiac function in a mouse model.
  • the levels of CRISPR/Cas9 activity may be sufficient for therapeutic benefit.
  • HEK293T cells which is an immortalized and aneuploid cell line that expresses very high levels of Cas9 and sgRNA, did not occur at as high a level and in some cases were undetectable in the hDMD myoblasts.
  • this level of specificity may be tolerable given the severity of DMD, the lack of an apparent cytotoxic effect in human cells
  • An engineered AAV capsid termed SASTG (Fig. 34; SEQ ID NOs: 436 and 437), was developed for enhanced cardiac and skeletal muscle tissue tropism (Piacentino et al. (2012) Human Gene Therapy 23:635-646).
  • a ZFN targeting the Rosa26 locus (“Rosa26 ZFN"; Fig. 33; SEQ ID NO: 434 and 435) was shown to be highly active in mouse cells (Perez -Pinera et al. Nucleic Acids Research (2012) 40:3741-3752).
  • AAV-SASTG vectors encoding the Rosa26 ZFN protein were designed and subsequently generated and purified by the UNC Viral Vector Core.
  • the Surveyor assay (Guschin et al, Methods Mol Biol 649, 247-256 (2010)) was used to demonstrate NHEJ mutagenesis at the Rosa26 locus following delivery of AAV-SASTG Rosa26 ZFNs in cultured C2C12 myoblasts that were actively cycling or forced into differentiation by serum removal (not shown).
  • AAV-SASTG vectors encoding the Rosa26 ZFN were injected directly into the tibialis anterior (TA) muscle of 6 week old C57BL6/J mice at titers of lelO vector genomes (vg) or 2.5el0 vg per muscle. Mice were sacrificed 4 weeks after injection and the TA muscles were harvested and partitioned into several fragments for genomic DNA extraction and analysis (Fig. 31). Genomic DNA was PCR amplified and subjected to the Surveyor assay to detect NHEJ mutations characteristic of ZFN mutagenesis at the Rosa26 target site (Fig. 32). Fig.
  • FIG. 32 shows Surveyor analysis of Rosa26 ZFN activities in skeletal muscle in vitro and in vivo following delivery of AAV-SASTG-ROSA.
  • Proliferating C2C12s were transduced with the indicated amount of virus and harvested at 4 days post-infection (Fig. 32(a)).
  • C2C12s were incubated in differentiation medium for 5 days and then transduced with the indicated amount of AAV-SASTG-ROSA virus in 24 well plates (Fig. 32(b)). Samples were collected at 10 days post-transduction.
  • the indicated amount of AAV-SASTG-ROSA was injected directly into the tibialis anterior of C57BL/6J mice and muscles were harvested 4 weeks post-infection.
  • the harvested TA muscles were partitioned into 8 separate pieces for genomic DNA analysis, each shown in a separate lane (Fig. 32(c)). Notably, high levels of gene modification were detected in all fragments at the highest dose (2.5el0 vg).
  • Example 15 shows Surveyor analysis of
  • AAV constructs are designed to therapeutically correct mutations of the dystrophin gene that cause Duchenne muscular dystrophy and skeletal and cardiac muscle degeneration.
  • CRISPR/Cas9 systems can be delivered using the AAV to restore the dystrophin reading frame by deleting exon 51, deleting exons 45-55, disrupting splice donor or acceptor sites, or creating frameshifts within exon 51 (Ousterout et al., Molecular Therapy 2013) to restore the dystrophin reading frame and protein expression.
  • the CRISPR/Cas9 system will include a Cas9 having a sequence of SEQ ID NO: 64 or 114 (See Figs. 40 and 41). gRNAs that could be combined with these Cas9s, targeting their respective PAM sequences, are provided (see Figs. 40 and 41; see also Tables 2 and 3).
  • iNs induced neurons
  • MEFs mouse embryonic fibroblasts
  • BAM factors BAM factors, Fig. 48
  • Other methods may include additional factors to induce various neuronal subtypes.
  • These experiments require transcription factors to be delivered ectopically, and the activation of the corresponding endogenous loci to sustain the neuronal phenotype.
  • the CRISPR/Cas9 system was engineered as a versatile transcription factor to activate endogenous genes in mammalian cells with the capacity to target any promoter in the genome through an RNA-guided mechanism (Figs. 49A,49B).
  • MEFs were seeded in either 24-well TCPS plates or on poly-D- lysine/laminin-coated coverslips. Following transduction of dCas9-VP64 and transfection of the gRNAs (see Tables 10 and 11 for sequences of gRNAs), the cells were cultured in MEF medium (Adler et al. Mol Ther Nucleic Acids 1 :e32 (2012)) for 24 hrs and then transferred to N3 neural induction medium (Vierbuchen et al. Nature 463 : 1035-1041 (2010)) for the duration of the experiment (Fig. 49B).
  • CR16-6 AS AAAC TCTTCTTCCCTCGGGCGCC (SEQ ID NO: 527) C 551
  • CR16-7 S cacc G GGGTGGGGGTACCAGAGGA (SEQ ID NO: 528) 552
  • CR16-7 AS AAAC TCCTCTGGTACCCCCACCC (SEQ ID NO: 529) C 553
  • CR16-8 S cacc G CCGGGGACAGAAGAGAGGG (SEQ ID NO: 530) 554
  • CR16-8 AS AAAC CCCTCTCTTCTGTCCCCGG (SEQ ID NO: 531) c 555
  • CR16-10 AS AAAC cgcatttgacagttacttt (SEQ ID NO: 535) c 559

Abstract

La présente invention concerne des compositions associées à un système à base de répétitions palindromiques courtes, groupées et régulièrement espacées (CRISPR) associées à Cas9 et des procédés d'utilisation desdites compositions associées audit système à base de CRISPR/Cas9 afin de modifier l'expression des gènes et dans le cadre du génie génomique. L'invention concerne également des compositions et des procédés d'utilisation desdites compositions afin de modifier l'expression des gènes et dans le cadre du génie génomique au niveau des muscles, par exemple des muscles squelettiques et cardiaque.
PCT/US2014/041190 2013-06-05 2014-06-05 Édition et régulation géniques à guidage arn WO2014197748A2 (fr)

Priority Applications (21)

Application Number Priority Date Filing Date Title
AU2014274840A AU2014274840B2 (en) 2013-06-05 2014-06-05 RNA-guided gene editing and gene regulation
EP18172956.7A EP3417880A1 (fr) 2013-06-05 2014-06-05 Édition et régulation géniques à guidage arn
KR1020237022139A KR20230107387A (ko) 2013-06-05 2014-06-05 Rna-가이드 유전자 편집 및 유전자 조절
US14/895,316 US10704060B2 (en) 2013-06-05 2014-06-05 RNA-guided gene editing and gene regulation
EP19168481.0A EP3539573B1 (fr) 2013-06-05 2014-06-05 Édition et régulation géniques à guidage arn
KR1020167000166A KR102307280B1 (ko) 2013-06-05 2014-06-05 Rna-가이드 유전자 편집 및 유전자 조절
JP2016518017A JP7085716B2 (ja) 2013-06-05 2014-06-05 Rnaガイド遺伝子編集及び遺伝子調節
CN201480044748.XA CN105658805B (zh) 2013-06-05 2014-06-05 Rna指导的基因编辑和基因调节
CN202111496852.0A CN114230675A (zh) 2013-06-05 2014-06-05 Rna指导的基因编辑和基因调节
KR1020237031879A KR20230136697A (ko) 2013-06-05 2014-06-05 Rna-가이드 유전자 편집 및 유전자 조절
KR1020217017616A KR102551324B1 (ko) 2013-06-05 2014-06-05 Rna-가이드 유전자 편집 및 유전자 조절
CA2914519A CA2914519A1 (fr) 2013-06-05 2014-06-05 Edition et regulation geniques a guidage arn
EP14806852.1A EP3004370A4 (fr) 2013-06-05 2014-06-05 Édition et régulation géniques à guidage arn
US15/991,333 US10745714B2 (en) 2013-06-05 2018-05-29 RNA-guided gene editing and gene regulation
US16/858,689 US20210002665A1 (en) 2013-06-05 2020-04-26 Rna-guided gene editing and gene regulation
AU2020203924A AU2020203924A1 (en) 2013-06-05 2020-06-12 RNA-guided gene editing and gene regulation
US16/927,679 US20210032654A1 (en) 2013-06-05 2020-07-13 Rna-guided gene editing and gene regulation
JP2022052186A JP2022101562A (ja) 2013-06-05 2022-03-28 Rnaガイド遺伝子編集及び遺伝子調節
AU2022250572A AU2022250572A1 (en) 2013-06-05 2022-10-10 RNA-guided gene editing and gene regulation
AU2022250450A AU2022250450A1 (en) 2013-06-05 2022-10-10 RNA-guided gene editing and gene regulation
JP2023131956A JP2023164823A (ja) 2013-06-05 2023-08-14 Rnaガイド遺伝子編集及び遺伝子調節

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US201361831481P 2013-06-05 2013-06-05
US61/831,481 2013-06-05
US201361839127P 2013-06-25 2013-06-25
US61/839,127 2013-06-25
US201361904911P 2013-11-15 2013-11-15
US61/904,911 2013-11-15
US201461967466P 2014-03-19 2014-03-19
US61/967,466 2014-03-19
US201461981575P 2014-04-18 2014-04-18
US61/981,575 2014-04-18

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US14/895,316 A-371-Of-International US10704060B2 (en) 2013-06-05 2014-06-05 RNA-guided gene editing and gene regulation
US15/991,333 Division US10745714B2 (en) 2013-06-05 2018-05-29 RNA-guided gene editing and gene regulation
US16/858,689 Continuation US20210002665A1 (en) 2013-06-05 2020-04-26 Rna-guided gene editing and gene regulation

Publications (2)

Publication Number Publication Date
WO2014197748A2 true WO2014197748A2 (fr) 2014-12-11
WO2014197748A3 WO2014197748A3 (fr) 2015-02-19

Family

ID=52008753

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/041190 WO2014197748A2 (fr) 2013-06-05 2014-06-05 Édition et régulation géniques à guidage arn

Country Status (8)

Country Link
US (4) US10704060B2 (fr)
EP (3) EP3539573B1 (fr)
JP (4) JP7085716B2 (fr)
KR (4) KR102551324B1 (fr)
CN (2) CN105658805B (fr)
AU (4) AU2014274840B2 (fr)
CA (1) CA2914519A1 (fr)
WO (1) WO2014197748A2 (fr)

Cited By (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015089462A1 (fr) * 2013-12-12 2015-06-18 The Broad Institute Inc. Distribution, utilisation et applications thérapeutiques des systèmes crispr-cas et compositions pour l'édition du génome
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
CN105255886A (zh) * 2015-07-09 2016-01-20 青岛市畜牧兽医研究所 一对靶向猪RelA基因的sgRNA
US20160058889A1 (en) * 2014-08-11 2016-03-03 The Board Of Regents Of The University Of Texas System Prevention of muscular dystrophy by crispr/cas9-mediated gene editing
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
WO2016049024A3 (fr) * 2014-09-24 2016-05-12 The Broad Institute Inc. Administration, utilisation et applications thérapeutiques des systèmes crispr-cas et compositions permettant de modéliser la concurrence de multiples mutations cancéreuses in vivo
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
JP2016103986A (ja) * 2014-12-01 2016-06-09 国立大学法人 東京大学 複数のユニットが多重に連結したdnaカセットおよび該カセットを含むベクターの製造方法
GB2534074A (en) * 2011-12-30 2016-07-13 Caribou Biosciences Inc Modified cascade ribonucleoproteins and uses thereof
WO2016131009A1 (fr) * 2015-02-13 2016-08-18 University Of Massachusetts Compositions et procédés pour l'administration transitoire de nucléases
WO2016130600A2 (fr) 2015-02-09 2016-08-18 Duke University Compositions et procédés pour l'édition de l'épigénome
EP3064587A1 (fr) 2015-03-06 2016-09-07 Leibniz-Institut für Pflanzenbiochemie (IPB) Bibliothèque de promoteurs synthétiques permettant de coordonner l'expression génique dans des cellules eucaryotes ou des organismes
WO2016161380A1 (fr) * 2015-04-01 2016-10-06 Editas Medicine, Inc. Méthodes et compositions liées à crispr/cas pour traiter la dystrophie musculaire de duchenne et la dystrophie musculaire de becker
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
WO2017015637A1 (fr) * 2015-07-22 2017-01-26 Duke University Criblage à haut rendement d'une fonction d'élément de régulation à l'aide de technologies d'édition de l'épigénome
WO2016205688A3 (fr) * 2015-06-18 2017-02-02 Bowles Robert D Régulation de la transcription à guidage arn et procédés d'utilisation de celle-ci pour le traitement des lombalgies
WO2017043647A1 (fr) * 2015-09-11 2017-03-16 国立研究開発法人理化学研究所 Procédé de déméthylation de l'adn spécifique à un site de liaison du facteur de transcription
US9596835B2 (en) 2008-05-28 2017-03-21 University Of Massachusetts Isolation of novel AAV's and uses thereof
WO2017035416A3 (fr) * 2015-08-25 2017-04-06 Duke University Compositions et procédés d'amélioration de la spécificité dans l'ingénierie génomique à l'aide d'endonucléases guidées par arn
WO2017072590A1 (fr) * 2015-10-28 2017-05-04 Crispr Therapeutics Ag Matériaux et méthodes pour traiter la dystrophie musculaire de duchenne
WO2017075335A1 (fr) 2015-10-28 2017-05-04 Voyager Therapeutics, Inc. Expression régulable au moyen d'un virus adéno-associé (vaa)
JP2017512767A (ja) * 2014-03-12 2017-05-25 プレシジョン バイオサイエンシズ,インク. 改変ヌクレアーゼを用いたジストロフィン遺伝子エクソンの欠失
WO2017066497A3 (fr) * 2015-10-13 2017-06-29 Duke University Ingénierie génomique avec systèmes crispr de type i dans des cellules eucaryotes
WO2017123910A1 (fr) * 2016-01-14 2017-07-20 The Brigham And Women's Hospital, Inc. Édition génomique pour traiter un glioblastome
CN107012158A (zh) * 2017-03-28 2017-08-04 东南大学 一种端粒酶启动基因表达方法及其应用
WO2017139505A2 (fr) 2016-02-11 2017-08-17 The Regents Of The University Of California Procédés et compositions pour modifier un gène mutant de la dystrophine dans le génome d'une cellule
CN107058358A (zh) * 2017-04-01 2017-08-18 中国科学院微生物研究所 一种双spacer序列识别切割CRISPR‑Cas9载体构建及其在疣孢菌中的应用
EP3219799A1 (fr) 2016-03-17 2017-09-20 IMBA-Institut für Molekulare Biotechnologie GmbH Expression sgrna crispr conditionnelle
WO2017160890A1 (fr) * 2016-03-14 2017-09-21 Editas Medicine, Inc. Méthodes et compositions liées à crispr/cas pour traiter les bêta-hémoglobinpathies
WO2017193029A2 (fr) 2016-05-05 2017-11-09 Duke University Méthodes liées à crispr/cas et compositions destinées à traiter la dystrophie musculaire de duchenne
WO2017180915A3 (fr) * 2016-04-13 2017-11-23 Duke University Répresseurs à base de crispr/cas9 pour inactiver des cibles géniques in vivo et procédés d'utilisation
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
WO2018009534A1 (fr) * 2016-07-05 2018-01-11 The Johns Hopkins University Compositions et procédés comprenant des améliorations d'arn guides de crispr à l'aide du promoteur h1
US9885057B2 (en) 2011-04-21 2018-02-06 University Of Massachusetts RAAV-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
GB2552861A (en) * 2016-06-02 2018-02-14 Sigma Aldrich Co Llc Using programmable DNA binding proteins to enhance targeted genome modification
KR20180034402A (ko) * 2015-06-18 2018-04-04 더 브로드 인스티튜트, 인코퍼레이티드 신규한 crispr 효소 및 시스템
US9938521B2 (en) 2014-03-10 2018-04-10 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating leber's congenital amaurosis 10 (LCA10)
WO2018078301A1 (fr) * 2016-10-28 2018-05-03 Université Pierre Et Marie Curie - Paris 6 (Upmc) Procédé de différenciation de cellules souches pluripotentes en cardiomyocytes
WO2018081978A1 (fr) * 2016-11-03 2018-05-11 深圳华大基因研究院 Méthode et système d'amélioration de l'efficacité de l'édition de gènes
WO2018098480A1 (fr) * 2016-11-28 2018-05-31 The Board Of Regents Of The University Of Texas System Prévention de la dystrophie musculaire par édition de gène médiée par crispr/cpf1
EP3239298A4 (fr) * 2014-12-26 2018-06-13 Riken Procédé d'invalidation génique
WO2018129296A1 (fr) * 2017-01-05 2018-07-12 The Board Of Regents Of The University Of Texas System Stratégie optimisée pour des modifications par saut d'exon à l'aide de crispr/cas9 avec des séquences de guidage triple
US10035825B2 (en) 2009-05-28 2018-07-31 University Of Massachusetts AAV's and uses thereof
WO2018147343A1 (fr) * 2017-02-07 2018-08-16 Edigene Corporation Méthode de traitement de maladies associées à une expression élevée de kras au moyen d'un système crispr-gndm
WO2018156824A1 (fr) * 2017-02-23 2018-08-30 President And Fellows Of Harvard College Méthodes de modification génétique d'une cellule
US10072251B2 (en) 2014-02-19 2018-09-11 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
WO2018170184A1 (fr) * 2017-03-14 2018-09-20 Editas Medicine, Inc. Systèmes et méthodes pour le traitement d'hémoglobinopathies
EP3385373A1 (fr) 2017-04-05 2018-10-10 Centro de Neurociências e Biologia Celular Compositions pour la reprogrammation de cellules en cellules dendritiques ou en cellules présentatrices d'antigènes, leurs procédés et utilisations
WO2018185709A1 (fr) 2017-04-05 2018-10-11 Centro De Neurociencias E Biologia Celular Compositions destinées à la reprogrammation de cellules en cellules dendritiques ou en cellules présentatrices d'antigène, procédés et utilisations associés
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
CN108779466A (zh) * 2015-11-30 2018-11-09 杜克大学 用于通过基因编辑修正人肌营养不良蛋白基因的治疗靶标和使用方法
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2019003193A1 (fr) * 2017-06-30 2019-01-03 Novartis Ag Méthodes pour le traitement d'une maladie à l'aide de systèmes d'édition de gènes
WO2018209158A3 (fr) * 2017-05-10 2019-01-03 Editas Medicine, Inc. Systèmes et procédés de nucléase guidé par crispr/arn
EP3332008A4 (fr) * 2015-09-23 2019-01-16 Université Laval Modification du gène de la dystrophine et ses utilisations
US10280418B2 (en) 2014-03-18 2019-05-07 Univeristy Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
WO2019131829A1 (fr) 2017-12-28 2019-07-04 国立大学法人京都大学 Composition de modification de gène cible
US10370432B2 (en) 2014-10-03 2019-08-06 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
US10480011B2 (en) 2014-10-21 2019-11-19 University Of Massachusetts Recombinant AAV variants and uses thereof
US10494621B2 (en) 2015-06-18 2019-12-03 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US10550372B2 (en) 2013-12-12 2020-02-04 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
WO2020030783A2 (fr) 2018-08-10 2020-02-13 Vib Vzw Moyens et procédés de tolérance à la sécheresse dans des cultures
US10577630B2 (en) 2013-06-17 2020-03-03 The Broad Institute, Inc. Delivery and use of the CRISPR-Cas systems, vectors and compositions for hepatic targeting and therapy
WO2020092725A1 (fr) * 2018-11-01 2020-05-07 Montana State University Modulation génique dotée d'un système crispr de type i
WO2020101042A1 (fr) * 2018-11-16 2020-05-22 Astellas Pharma Inc. Méthode de traitement de la dystrophie musculaire par ciblage du gène de l'utrophine
US10687520B2 (en) 2017-03-07 2020-06-23 The Board Of Regents Of The University Of Texas System Generation and correction of a humanized mouse model with a deletion of dystrophin exon 44
EP3553090A4 (fr) * 2016-12-12 2020-06-24 Astellas Pharma Inc. Polypeptide fusionné à régulation transcriptionnelle
US10696986B2 (en) 2014-12-12 2020-06-30 The Board Institute, Inc. Protected guide RNAS (PGRNAS)
US10711270B2 (en) 2014-10-03 2020-07-14 University Of Massachusetts High efficiency library-identified AAV vectors
US10711285B2 (en) 2013-06-17 2020-07-14 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US10738305B2 (en) 2015-02-23 2020-08-11 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US10781444B2 (en) 2013-06-17 2020-09-22 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US20200340012A1 (en) * 2016-08-18 2020-10-29 The Regents Of The University Of California Crispr-cas genome engineering via a modular aav delivery system
WO2020209800A3 (fr) * 2019-04-11 2020-11-12 Agency For Science, Technology And Research Procédé de modification d'un état de différenciation d'une cellule
WO2020237237A1 (fr) * 2019-05-23 2020-11-26 American Molecular Laboratories, Inc. Procédés de détection d'un niveau de h.pylori dans un échantillon fécal
US10851357B2 (en) 2013-12-12 2020-12-01 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
EP3596117A4 (fr) * 2017-03-17 2021-01-13 The Johns Hopkins University Thérapie épigénétique ciblée contre l'élément de régulation distale d'expression du tgfb2
US10930367B2 (en) 2012-12-12 2021-02-23 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and conveying results thereof
US10946108B2 (en) 2013-06-17 2021-03-16 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
WO2021051054A1 (fr) * 2019-09-13 2021-03-18 The Regents Of The University Of Colorado, A Body Corporate Procédés et compositions pour rajeunissement cellulaire et tissulaire
CN112534054A (zh) * 2018-05-11 2021-03-19 比姆医疗股份有限公司 使用可编程碱基编辑器系统取代病原性氨基酸的方法
WO2021058543A1 (fr) * 2019-09-23 2021-04-01 Michalakis Stylianos Procédé de transactivation d'un gène homologue d'un gène d'intérêt et procédé in vitro de diagnostic d'une maladie
US10975391B2 (en) 2014-04-25 2021-04-13 University Of Massachusetts Recombinant AAV vectors useful for reducing immunity against transgene products
WO2021089736A1 (fr) 2019-11-06 2021-05-14 Association Institut De Myologie Polythérapie pour maladies musculaires
US11008588B2 (en) 2013-06-17 2021-05-18 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
US11028388B2 (en) 2014-03-05 2021-06-08 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for treating Usher syndrome and retinitis pigmentosa
US11041173B2 (en) 2012-12-12 2021-06-22 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
US11060088B2 (en) 2016-02-12 2021-07-13 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11155795B2 (en) 2013-12-12 2021-10-26 The Broad Institute, Inc. CRISPR-Cas systems, crystal structure and uses thereof
CN113637658A (zh) * 2021-08-19 2021-11-12 中国农业科学院棉花研究所 基于dCas9-oToV的基因转录系统及其应用
US11180793B2 (en) 2015-04-24 2021-11-23 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
CN113785063A (zh) * 2019-04-14 2021-12-10 杜克大学 用于治疗杜氏肌营养不良症的aav载体介导的大规模突变热点缺失
US11207426B2 (en) 2016-04-05 2021-12-28 University Of Massachusetts Compositions and methods for selective inhibition of grainyhead-like protein expression
WO2022008557A2 (fr) 2020-07-08 2022-01-13 UCB Biopharma SRL Modulation de l'expression de cftr
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
US11253576B2 (en) 2015-10-22 2022-02-22 University Of Massachusetts Methods and compositions for treating metabolic imbalance in neurodegenerative disease
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11268077B2 (en) 2018-02-05 2022-03-08 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
EP3836974A4 (fr) * 2018-08-18 2022-06-15 President and Fellows of Harvard College Édition génique in situ
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11413356B2 (en) 2016-04-15 2022-08-16 University Of Massachusetts Methods and compositions for treating metabolic imbalance
US11426469B2 (en) 2015-10-22 2022-08-30 University Of Massachusetts Prostate-targeting adeno-associated virus serotype vectors
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11479761B2 (en) 2015-04-16 2022-10-25 Wageningen Universiteit Nuclease-mediated genome editing
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
WO2023010133A2 (fr) 2021-07-30 2023-02-02 Tune Therapeutics, Inc. Compositions et procédés de modulation de l'expression de la frataxine
WO2023010135A1 (fr) 2021-07-30 2023-02-02 Tune Therapeutics, Inc. Compositions et procédés pour moduler l'expression de la protéine 2 de liaison méthyle-cpg (mecp2)
US11572574B2 (en) 2017-09-28 2023-02-07 Toolgen Incorporated Artificial genome manipulation for gene expression regulation
US11578340B2 (en) 2016-10-13 2023-02-14 University Of Massachusetts AAV capsid designs
US11578312B2 (en) 2015-06-18 2023-02-14 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of CAS9 orthologs and variants for sequence manipulation
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11634755B2 (en) 2015-06-18 2023-04-25 The Broad Institute, Inc. Crispr enzymes and systems
US11661583B2 (en) 2015-08-27 2023-05-30 University Of Washington Drug discovery platform for Duchenne cardiomyopathy
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11666665B2 (en) 2014-12-01 2023-06-06 President And Fellows Of Harvard College RNA-guided systems for in vivo gene editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
WO2023137472A2 (fr) 2022-01-14 2023-07-20 Tune Therapeutics, Inc. Compositions, systèmes et procédés de programmation de phénotypes de lymphocytes t par répression génique ciblée
WO2023137471A1 (fr) 2022-01-14 2023-07-20 Tune Therapeutics, Inc. Compositions, systèmes et procédés de programmation de phénotypes de lymphocytes t par activation génique ciblée
EP4215614A1 (fr) 2022-01-24 2023-07-26 Dynacure Thérapie de combinaison pour maladies liées à la dystrophine
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11739330B2 (en) 2017-09-22 2023-08-29 University Of Massachusetts SOD1 dual expression vectors and uses thereof
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11826433B2 (en) 2016-02-02 2023-11-28 University Of Massachusetts Method to enhance the efficiency of systemic AAV gene delivery to the central nervous system
WO2023247789A1 (fr) 2022-06-24 2023-12-28 European Molecular Biology Laboratory Outil modulaire basé sur une crispr pour l'introduction spécifique de modifications épigénétiques sur des loci cibles
WO2023250511A2 (fr) 2022-06-24 2023-12-28 Tune Therapeutics, Inc. Compositions, systèmes et procédés de réduction de lipoprotéine de faible densité par répression génique ciblée
US11859179B2 (en) 2017-05-09 2024-01-02 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (ALS)
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
WO2024015881A2 (fr) 2022-07-12 2024-01-18 Tune Therapeutics, Inc. Compositions, systèmes et procédés d'activation transcriptionnelle ciblée
US11882815B2 (en) 2016-06-15 2024-01-30 University Of Massachusetts Recombinant adeno-associated viruses for delivering gene editing molecules to embryonic cells
US11891603B2 (en) 2017-07-21 2024-02-06 The Governors Of The University Of Alberta Antisense oligonucleotides that bind to exon 51 of human dystrophin pre-mRNA
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
WO2024040254A2 (fr) 2022-08-19 2024-02-22 Tune Therapeutics, Inc. Compositions, systèmes et méthodes de régulation du virus de l'hépatite b par répression génique ciblée
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US11920128B2 (en) 2013-09-18 2024-03-05 Kymab Limited Methods, cells and organisms
WO2024064642A2 (fr) 2022-09-19 2024-03-28 Tune Therapeutics, Inc. Compositions, systèmes et méthodes de modulation de fonction de lymphocyte t
KR102655989B1 (ko) * 2017-07-21 2024-04-08 더 거버너스 오브 더 유니버시티 오브 알버타 인간 디스트로핀 pre-mRNA의 엑손 51에 결합하는 안티센스 올리고뉴클레오티드

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9737480B2 (en) 2012-02-06 2017-08-22 President And Fellows Of Harvard College ARRDC1-mediated microvesicles (ARMMs) and uses thereof
WO2014071219A1 (fr) * 2012-11-01 2014-05-08 Factor Bioscience Inc. Procédés et produits pour l'expression de protéines dans des cellules
AU2014274840B2 (en) * 2013-06-05 2020-03-12 Duke University RNA-guided gene editing and gene regulation
US9816080B2 (en) 2014-10-31 2017-11-14 President And Fellows Of Harvard College Delivery of CAS9 via ARRDC1-mediated microvesicles (ARMMs)
AU2015373893B2 (en) * 2014-12-31 2021-07-29 Synthetic Genomics, Inc. Compositions and methods for high efficiency in vivo genome editing
US11674144B2 (en) * 2015-04-16 2023-06-13 California Institute Of Technology Fractional regulation of transcription
WO2017053729A1 (fr) 2015-09-25 2017-03-30 The Board Of Trustees Of The Leland Stanford Junior University Édition du génome à médiation par une nucléase de cellules primaires et leur enrichissement
EP3359661A4 (fr) 2015-10-08 2019-05-15 President and Fellows of Harvard College Édition multiplexée de génome
EA201891614A1 (ru) 2016-01-11 2019-02-28 Те Борд Оф Трастиз Оф Те Лилэнд Стэнфорд Джуниор Юниверсити Химерные белки и способы иммунотерапии
EA201891619A1 (ru) 2016-01-11 2019-02-28 Те Борд Оф Трастиз Оф Те Лилэнд Стэнфорд Джуниор Юниверсити Химерные белки и способы регулирования экспрессии генов
CN106119269B (zh) * 2016-06-23 2019-12-06 百奥迈科生物技术有限公司 一种在大肠杆菌中制备线性单链dna的方法
EP3497221A4 (fr) * 2016-08-10 2020-02-05 Duke University Compositions, systèmes et procédés de programmation d'une fonction de cellules immunitaires par régulation ciblée de gènes
RU2019103691A (ru) * 2016-08-19 2020-09-22 Тулджен Инкорпорейтед Искусственно сконструированная система регуляции ангиогенеза
CN107880132B (zh) * 2016-09-30 2022-06-17 北京大学 一种融合蛋白及使用其进行同源重组的方法
US11730823B2 (en) 2016-10-03 2023-08-22 President And Fellows Of Harvard College Delivery of therapeutic RNAs via ARRDC1-mediated microvesicles
CN106318947A (zh) * 2016-10-17 2017-01-11 北京大北农科技集团股份有限公司 基因组编辑系统及其用途
CN110214184A (zh) 2016-12-01 2019-09-06 桑格摩生物治疗股份有限公司 τ蛋白调节剂以及用于其递送的方法和组合物
CA3049989A1 (fr) 2017-02-10 2018-08-16 Zymergen Inc. Strategie de conception de plasmide universelle modulaire pour l'assemblage et l'edition de multiples constructions d'adn pour hotes multiples
US10801038B2 (en) * 2017-02-28 2020-10-13 Trustees Of Boston University Opto-genetic modulator
WO2018169983A1 (fr) * 2017-03-13 2018-09-20 President And Fellows Of Harvard College Procédés de modulation de l'expression de séquences d'acides nucléiques cibles dans une cellule
EP3617311A4 (fr) * 2017-03-30 2021-04-21 Kyoto University Procédé pour induire un saut d'exon par édition génomique
EP3635120A1 (fr) * 2017-06-02 2020-04-15 Institut National de la Santé et de la Recherche Médicale (INSERM) Vecteur lentiviral recombinant pour la thérapie génique de la drépanocytose à base de cellules souches
AU2018283372A1 (en) 2017-06-16 2020-02-06 Avery Therapeutics, Inc. Three dimensional tissue compositions and methods of use
CN107488649A (zh) * 2017-08-25 2017-12-19 南方医科大学 一种Cpf1和p300核心结构域的融合蛋白、相应的DNA靶向激活系统和应用
WO2019055536A1 (fr) * 2017-09-14 2019-03-21 The Children's Medical Center Corporation Compositions et méthodes permettant d'améliorer l'efficacité d'une transplantation de cellules
CN107513531B (zh) * 2017-09-21 2020-02-21 无锡市妇幼保健院 用于内源性过表达lncRNA-XIST的gRNA靶点序列及其应用
CN107722125B (zh) * 2017-09-28 2021-05-07 中山大学 一种人工转录激活因子dCas9-TV及其编码基因与应用
WO2019089623A1 (fr) * 2017-10-30 2019-05-09 Children's Hospital Medical Center Protéines de fusion destinées à être utilisées pour améliorer la correction génique par recombinaison homologue
US20200354419A1 (en) * 2017-11-03 2020-11-12 Hunterian Medicine Llc Compositions and methods of use thereof for the treatment of duchenne muscular dystrophy
KR102132198B1 (ko) * 2017-12-14 2020-07-09 단국대학교 산학협력단 Tert 프로모터 돌연변이에 특이적인 crispr 시스템 및 그의 이용
CN108559731A (zh) * 2018-01-15 2018-09-21 南方医科大学 一种可调控基因表达的人类胚胎干细胞系及其应用
CN110229814A (zh) * 2018-03-06 2019-09-13 中国科学院动物研究所 改进的向导rna
AU2019255789A1 (en) 2018-04-19 2020-10-22 The Regents Of The University Of California Compositions and methods for gene editing
CN112166194A (zh) * 2018-04-23 2021-01-01 杜克大学 通过dna甲基化的靶向编辑下调snca的表达
WO2019210279A1 (fr) * 2018-04-27 2019-10-31 The Regents Of The University Of California Formation de novo du système biliaire par transdifférenciation des hépatocytes
GB2589246A (en) 2018-05-16 2021-05-26 Synthego Corp Methods and systems for guide RNA design and use
WO2020010249A1 (fr) * 2018-07-06 2020-01-09 The Regents Of The University Of California Nouveau procédé d'ingénierie de tissus humains translantables
CN109055375B (zh) * 2018-07-26 2021-11-12 东南大学 一种crispr辅助反式增强子激活基因表达的方法及其应用
CN110760511B (zh) * 2018-07-27 2021-09-07 广东赤萌医疗科技有限公司 一种用于治疗杜氏肌营养不良症的gRNA、表达载体、CRISPR-Cas9系统
JP2021532831A (ja) 2018-08-02 2021-12-02 ダイン セラピューティクス, インコーポレーテッドDyne Therapeutics, Inc. ジストロフィン異常症を処置するための筋標的化複合体およびそれらの使用
US11168141B2 (en) 2018-08-02 2021-11-09 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
CA3108289A1 (fr) 2018-08-02 2020-02-06 Dyne Therapeutics, Inc. Complexes de ciblage musculaire et leurs utilisations pour le traitement de la dystrophie musculaire facio-scapulo-humerale
CA3107002A1 (fr) 2018-08-15 2020-04-30 Zymergen Inc. Applications de crispri dans l'ingenierie metabolique a haut rendement
EP3864161A4 (fr) * 2018-10-09 2022-11-23 The University of North Carolina at Chapel Hill Système d'édition de gènes régulé
US20210386827A1 (en) * 2018-10-15 2021-12-16 Avery Therapeutics, Inc. Cell-free compositions and methods for restoration or enhancement of tissue function
US20210403942A1 (en) * 2018-11-06 2021-12-30 Cellino Biotech, Inc. Systems for cell control
WO2020159880A1 (fr) * 2019-01-28 2020-08-06 The Administrators Of The Tulane Educational Fund Procédés à base de crispr et nouvelles compositions pour le traitement de troubles vasculaires
WO2020168133A1 (fr) 2019-02-13 2020-08-20 Beam Therapeutics Inc. Compositions et méthodes de traitement d'hémoglobinopathies
CN109929865B (zh) * 2019-03-11 2022-03-08 东南大学 基于gal4-uas系统的crispr辅助反式增强子激活基因表达的方法及其应用
EP3952925A4 (fr) * 2019-04-12 2024-01-24 Univ California Compositions et méthodes pour modifier des gènes de la dystrophine
WO2020214613A1 (fr) * 2019-04-14 2020-10-22 Duke University Composition d'édition génomique à base de crispr/cas permettant de restaurer la fonction de la dystrophine
WO2020232373A1 (fr) * 2019-05-15 2020-11-19 Board Of Regents, The University Of Texas System Méthodes crispr pour le traitement de cancers
CN112266418A (zh) * 2019-07-08 2021-01-26 中国科学院微生物研究所 改进的基因组编辑系统及其应用
WO2021011936A2 (fr) 2019-07-18 2021-01-21 University Of Rochester Immunoprotection sélective de type cellulaire de cellules
US20230272428A1 (en) * 2019-12-16 2023-08-31 President And Fellows Of Harvard College Methods and compositions for correction of dmd mutations
JP2023515709A (ja) * 2020-04-27 2023-04-13 デューク ユニバーシティ 筋肉特異的プロモーターをコードするaavベクターを使用するインビボでの衛星細胞の遺伝子編集
WO2021222327A1 (fr) * 2020-04-27 2021-11-04 Duke University Procédé de criblage à haut rendement pour découvrir des paires de grna optimales pour une délétion d'exon médiée par crispr
EP4149622A1 (fr) * 2020-05-12 2023-03-22 Myogene Bio LLC Agents immunosuppresseurs et méthodes de re-dosage d'administration virale pour thérapie génique
CN112063621B (zh) * 2020-09-02 2022-06-28 西湖大学 杜氏肌营养不良症相关的外显子剪接增强子、sgRNA、基因编辑工具及应用
WO2022060841A2 (fr) 2020-09-15 2022-03-24 Research Institute At Nationwide Children's Hospital Édition de gène d'intégration ciblée indépendante de l'homologie médiée par vaa pour la correction de diverses mutations dmd chez des patients atteints d'une dystrophie musculaire
CN116802289A (zh) * 2020-10-15 2023-09-22 东北大学 用于增加胶原蛋白产量的工程化细胞
JP2023549456A (ja) * 2020-10-21 2023-11-27 デューク ユニバーシティ デュシェンヌ型筋ジストロフィーの処置のための大きい変異ホットスポットの二重aavベクター媒介欠失
CN112662674B (zh) * 2021-01-12 2023-04-11 广州瑞风生物科技有限公司 靶向编辑VEGFA基因外显子区域的gRNA及其应用
CN114854791A (zh) * 2021-02-04 2022-08-05 北京中因科技有限公司 一种新型CRISPR-Cas9系统载体及其应用
CN112779259B (zh) * 2021-03-18 2023-05-26 新疆畜牧科学院生物技术研究所(新疆畜牧科学院中国-澳大利亚绵羊育种研究中心) 一种用于精准编辑绵羊OCT4基因的sgRNA、扩增用引物和应用
US11771776B2 (en) 2021-07-09 2023-10-03 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
US11638761B2 (en) 2021-07-09 2023-05-02 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating Facioscapulohumeral muscular dystrophy
WO2023039346A1 (fr) * 2021-09-10 2023-03-16 The Regents Of The University Of California Systèmes crispr-cas régulés en température et leurs procédés d'utilisation
WO2023050158A1 (fr) * 2021-09-29 2023-04-06 深圳先进技术研究院 Procédé pour réaliser une édition sur plusieurs bases
WO2023212594A2 (fr) * 2022-04-26 2023-11-02 University Of Massachusetts Insertions de grande taille médiées par un arnpeg unique

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9206016D0 (en) 1992-03-19 1992-04-29 Sandoz Ltd Improvements in or relating to organic compounds
WO1993024640A2 (fr) 1992-06-04 1993-12-09 The Regents Of The University Of California PROCEDES ET COMPOSITIONS UTILISES DANS UNE THERAPIE GENIQUE $i(IN VIVO)
US5593972A (en) 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
EP0681483B1 (fr) 1993-01-26 2005-08-24 The Trustees of the University of Pennsylvania (a corporation of Pennsylvania) Compositions et procedes d'administration de matieres genetiques
US5962428A (en) 1995-03-30 1999-10-05 Apollon, Inc. Compositions and methods for delivery of genetic material
AU2001253812A1 (en) 2000-04-28 2001-11-12 Genzyme Corporation In vivo loading of mhc
AU2003295366B2 (en) 2002-11-04 2011-11-24 Advisys, Inc. Synthetic muscle promoters with activities exceeding naturally occurring regulatory sequences in cardiac cells
CN1759182A (zh) * 2002-11-22 2006-04-12 克雷顿研究院 调节基因的组合物和系统
AU2004263884A1 (en) 2003-08-08 2005-02-17 President And Fellows Of Harvard College SiRNA based methods for treating Alzheimer's disease
US7807816B2 (en) * 2004-06-28 2010-10-05 University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
WO2008006028A2 (fr) 2006-07-05 2008-01-10 The Scripps Research Institute Recombinases chimères à doigts de zinc optimisées pour catalyse par évolution directe
CN101896186A (zh) * 2007-10-26 2010-11-24 莱顿教学医院 对抗肌肉病症的方式和方法
EP2998402A1 (fr) 2008-10-17 2016-03-23 Joule Unlimited Technologies, Inc. Production d'éthanol par des microorganismes
EP2206723A1 (fr) 2009-01-12 2010-07-14 Bonas, Ulla Domaines modulaires de liaison à l'ADN
EP2480659A2 (fr) 2009-09-24 2012-08-01 Cellectis Réactifs à base de méganucléase et leurs utilisations pour traiter des maladies génétiques provoquées par des mutations de changement de phase/ non sens
BR122019025207B1 (pt) * 2009-10-22 2022-10-04 Sangamo Biosciences, Inc. Proteína de dedo de zinco de ocorrência não natural e proteína de fusão
BR112012020257A8 (pt) 2010-02-11 2018-02-14 Recombinetics Inc métodos e aparelhos para produzir artiodátilos transgênicos
EP2569424A1 (fr) 2010-05-12 2013-03-20 Cellectis Variants de méganucléase clivant une séquence cible d'adn du gène de dystrophine et leurs utilisations
WO2011146121A1 (fr) 2010-05-17 2011-11-24 Sangamo Biosciences, Inc. Nouvelles protéines se liant à l'adn et leurs utilisations
IT1400425B1 (it) 2010-06-08 2013-05-31 Amsterdam Molecular Therapeutics Bv Modified snrnas for use in therapy.
CN102643852B (zh) * 2011-02-28 2015-04-08 华东理工大学 光可控的基因表达系统
US9540623B2 (en) 2011-07-08 2017-01-10 Cellectis Method for increasing the efficiency of double-strand-break induced mutagenesis
CN104203289B (zh) * 2012-01-27 2020-11-03 比奥马林技术公司 用于治疗杜兴型肌营养不良症和贝克型肌营养不良症的具有改善特性的rna调节性寡核苷酸
WO2013152220A2 (fr) 2012-04-04 2013-10-10 Life Technologies Corporation Plate-forme d'assemblage d'effecteur tal, services personnalisés, kits et tests
EP2841572B1 (fr) 2012-04-27 2019-06-19 Duke University Correction génétique de gènes ayant subi une mutation
US9738879B2 (en) 2012-04-27 2017-08-22 Duke University Genetic correction of mutated genes
US9890364B2 (en) 2012-05-29 2018-02-13 The General Hospital Corporation TAL-Tet1 fusion proteins and methods of use thereof
US8884771B2 (en) * 2012-08-01 2014-11-11 Microchip Technology Incorporated Smoke detection using change in permittivity of capacitor air dielectric
RS60838B1 (sr) * 2012-08-29 2020-10-30 Sangamo Therapeutics Inc Postupci i kompozicije za tretman genskog stanja
WO2014071219A1 (fr) * 2012-11-01 2014-05-08 Factor Bioscience Inc. Procédés et produits pour l'expression de protéines dans des cellules
WO2014081855A1 (fr) 2012-11-20 2014-05-30 Universite De Montreal Procédés et compositions pour des dystrophies musculaires
KR102006880B1 (ko) * 2012-12-06 2019-08-02 시그마-알드리치 컴퍼니., 엘엘씨 Crispr-기초된 유전체 변형과 조절
EP4286402A3 (fr) 2012-12-12 2024-02-14 The Broad Institute, Inc. Systèmes de composants crispr-cas, procédés et compositions pour la manipulation de séquence
US8697359B1 (en) * 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
CA2906869C (fr) 2013-03-15 2022-08-02 Edward J. Britt Dispositif de conversion energetique et son procede de fabrication et d'utilisation
EP2997146A4 (fr) * 2013-05-15 2017-04-26 Sangamo BioSciences, Inc. Procédés et compositions pour le traitement d'une maladie génétique
US9267135B2 (en) 2013-06-04 2016-02-23 President And Fellows Of Harvard College RNA-guided transcriptional regulation
AU2014274840B2 (en) * 2013-06-05 2020-03-12 Duke University RNA-guided gene editing and gene regulation
EP3011032B1 (fr) * 2013-06-17 2019-10-16 The Broad Institute, Inc. Délivrance, modification et optimisation de systèmes, procédés et compositions pour cibler et modéliser des maladies et des troubles liés aux cellules post-mitotiques

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (303)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11826434B2 (en) 2008-05-28 2023-11-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US9596835B2 (en) 2008-05-28 2017-03-21 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10166297B2 (en) 2008-05-28 2019-01-01 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10300146B2 (en) 2008-05-28 2019-05-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10905776B2 (en) 2008-05-28 2021-02-02 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10689420B2 (en) 2009-05-28 2020-06-23 University Of Massachusetts AAV's and uses thereof
US10035825B2 (en) 2009-05-28 2018-07-31 University Of Massachusetts AAV's and uses thereof
US11834474B2 (en) 2009-05-28 2023-12-05 University Of Massachusetts AAV's and uses thereof
US10077452B2 (en) 2011-04-21 2018-09-18 University Of Massachusetts rAAV-based compositions and methods
US11920133B2 (en) 2011-04-21 2024-03-05 University Of Massachusetts RAAV-based compositions and methods
US11254939B2 (en) 2011-04-21 2022-02-22 University Of Massachusetts RAAV-based compositions and methods
US9885057B2 (en) 2011-04-21 2018-02-06 University Of Massachusetts RAAV-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
US10597656B2 (en) 2011-04-21 2020-03-24 University Of Massachusetts RAAV-based compositions and methods
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
GB2534074A (en) * 2011-12-30 2016-07-13 Caribou Biosciences Inc Modified cascade ribonucleoproteins and uses thereof
US11041173B2 (en) 2012-12-12 2021-06-22 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US10930367B2 (en) 2012-12-12 2021-02-23 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and conveying results thereof
US10946108B2 (en) 2013-06-17 2021-03-16 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
US11597949B2 (en) 2013-06-17 2023-03-07 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US11008588B2 (en) 2013-06-17 2021-05-18 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
US10781444B2 (en) 2013-06-17 2020-09-22 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US10711285B2 (en) 2013-06-17 2020-07-14 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US10577630B2 (en) 2013-06-17 2020-03-03 The Broad Institute, Inc. Delivery and use of the CRISPR-Cas systems, vectors and compositions for hepatic targeting and therapy
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US10954548B2 (en) 2013-08-09 2021-03-23 President And Fellows Of Harvard College Nuclease profiling system
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US10227581B2 (en) 2013-08-22 2019-03-12 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US10682410B2 (en) 2013-09-06 2020-06-16 President And Fellows Of Harvard College Delivery system for functional nucleases
US10912833B2 (en) 2013-09-06 2021-02-09 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US11920128B2 (en) 2013-09-18 2024-03-05 Kymab Limited Methods, cells and organisms
US10640788B2 (en) 2013-11-07 2020-05-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAs
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US11390887B2 (en) 2013-11-07 2022-07-19 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US11155795B2 (en) 2013-12-12 2021-10-26 The Broad Institute, Inc. CRISPR-Cas systems, crystal structure and uses thereof
WO2015089462A1 (fr) * 2013-12-12 2015-06-18 The Broad Institute Inc. Distribution, utilisation et applications thérapeutiques des systèmes crispr-cas et compositions pour l'édition du génome
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
US10851357B2 (en) 2013-12-12 2020-12-01 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US10550372B2 (en) 2013-12-12 2020-02-04 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US11597919B2 (en) 2013-12-12 2023-03-07 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US11407985B2 (en) 2013-12-12 2022-08-09 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for genome editing
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US11124782B2 (en) 2013-12-12 2021-09-21 President And Fellows Of Harvard College Cas variants for gene editing
US10072251B2 (en) 2014-02-19 2018-09-11 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US10894949B2 (en) 2014-02-19 2021-01-19 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US11028388B2 (en) 2014-03-05 2021-06-08 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for treating Usher syndrome and retinitis pigmentosa
US11268086B2 (en) 2014-03-10 2022-03-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating Leber's Congenital Amaurosis 10 (LCA10)
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US9938521B2 (en) 2014-03-10 2018-04-10 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating leber's congenital amaurosis 10 (LCA10)
US10253312B2 (en) 2014-03-10 2019-04-09 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating Leber's Congenital Amaurosis 10 (LCA10)
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
EP3116533A4 (fr) * 2014-03-12 2018-04-18 Precision Biosciences, Inc. Délétion d'exon du gène codant la dystrophine au moyen de nucléases génétiquement modifiées
EP3858376A1 (fr) * 2014-03-12 2021-08-04 Precision Biosciences, Inc. Délétion d'exon du gène codant la dystrophine au moyen de nucléases génétiquement modifiées
JP2017512767A (ja) * 2014-03-12 2017-05-25 プレシジョン バイオサイエンシズ,インク. 改変ヌクレアーゼを用いたジストロフィン遺伝子エクソンの欠失
JP2021088563A (ja) * 2014-03-12 2021-06-10 プレシジョン バイオサイエンシズ,インク. 改変ヌクレアーゼを用いたジストロフィン遺伝子エクソンの欠失
US10711274B2 (en) 2014-03-18 2020-07-14 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US11760999B2 (en) 2014-03-18 2023-09-19 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10954518B2 (en) 2014-03-18 2021-03-23 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10280418B2 (en) 2014-03-18 2019-05-07 Univeristy Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10851375B2 (en) 2014-03-18 2020-12-01 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
US10975391B2 (en) 2014-04-25 2021-04-13 University Of Massachusetts Recombinant AAV vectors useful for reducing immunity against transgene products
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10704062B2 (en) 2014-07-30 2020-07-07 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
JP2021073304A (ja) * 2014-08-11 2021-05-13 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム Crispr/cas9媒介遺伝子編集による筋ジストロフィーの予防
CN106714845A (zh) * 2014-08-11 2017-05-24 得克萨斯州大学系统董事会 通过crispr/cas9介导的基因编辑预防肌营养不良
JP7197617B2 (ja) 2014-08-11 2022-12-27 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム Crispr/cas9媒介遺伝子編集による筋ジストロフィーの予防
US20160058889A1 (en) * 2014-08-11 2016-03-03 The Board Of Regents Of The University Of Texas System Prevention of muscular dystrophy by crispr/cas9-mediated gene editing
JP2017525696A (ja) * 2014-08-11 2017-09-07 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム Crispr/cas9媒介遺伝子編集による筋ジストロフィーの予防
WO2016049024A3 (fr) * 2014-09-24 2016-05-12 The Broad Institute Inc. Administration, utilisation et applications thérapeutiques des systèmes crispr-cas et compositions permettant de modéliser la concurrence de multiples mutations cancéreuses in vivo
US11124796B2 (en) 2014-09-24 2021-09-21 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for modeling competition of multiple cancer mutations in vivo
US10370432B2 (en) 2014-10-03 2019-08-06 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US10711270B2 (en) 2014-10-03 2020-07-14 University Of Massachusetts High efficiency library-identified AAV vectors
US11014976B2 (en) 2014-10-03 2021-05-25 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US11542525B2 (en) 2014-10-21 2023-01-03 University Of Massachusetts Recombinant AAV variants and uses thereof
US10480011B2 (en) 2014-10-21 2019-11-19 University Of Massachusetts Recombinant AAV variants and uses thereof
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US11666665B2 (en) 2014-12-01 2023-06-06 President And Fellows Of Harvard College RNA-guided systems for in vivo gene editing
JP2016103986A (ja) * 2014-12-01 2016-06-09 国立大学法人 東京大学 複数のユニットが多重に連結したdnaカセットおよび該カセットを含むベクターの製造方法
US10696986B2 (en) 2014-12-12 2020-06-30 The Board Institute, Inc. Protected guide RNAS (PGRNAS)
US11624078B2 (en) 2014-12-12 2023-04-11 The Broad Institute, Inc. Protected guide RNAS (pgRNAS)
US10863730B2 (en) 2014-12-26 2020-12-15 Riken Gene knockout method
EP3239298A4 (fr) * 2014-12-26 2018-06-13 Riken Procédé d'invalidation génique
JP2018505219A (ja) * 2015-02-09 2018-02-22 デューク ユニバーシティ エピゲノム編集のための組成物および方法
US10676726B2 (en) 2015-02-09 2020-06-09 Duke University Compositions and methods for epigenome editing
JP2021164463A (ja) * 2015-02-09 2021-10-14 デューク ユニバーシティ エピゲノム編集のための組成物および方法
WO2016130600A2 (fr) 2015-02-09 2016-08-18 Duke University Compositions et procédés pour l'édition de l'épigénome
WO2016130600A3 (fr) * 2015-02-09 2016-11-03 Duke University Compositions et procédés pour l'édition de l'épigénome
EP3256487A4 (fr) * 2015-02-09 2018-07-18 Duke University Compositions et procédés pour l'édition de l'épigénome
US11155796B2 (en) 2015-02-09 2021-10-26 Duke University Compositions and methods for epigenome editing
US11920168B2 (en) 2015-02-13 2024-03-05 University Of Massachusetts Compositions and methods for transient delivery of nucleases
US10584321B2 (en) 2015-02-13 2020-03-10 University Of Massachusetts Compositions and methods for transient delivery of nucleases
WO2016131009A1 (fr) * 2015-02-13 2016-08-18 University Of Massachusetts Compositions et procédés pour l'administration transitoire de nucléases
US10738305B2 (en) 2015-02-23 2020-08-11 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
EP3064587A1 (fr) 2015-03-06 2016-09-07 Leibniz-Institut für Pflanzenbiochemie (IPB) Bibliothèque de promoteurs synthétiques permettant de coordonner l'expression génique dans des cellules eucaryotes ou des organismes
WO2016161380A1 (fr) * 2015-04-01 2016-10-06 Editas Medicine, Inc. Méthodes et compositions liées à crispr/cas pour traiter la dystrophie musculaire de duchenne et la dystrophie musculaire de becker
EP3748004A1 (fr) * 2015-04-01 2020-12-09 Editas Medicine, Inc. Méthodes et compositions liées à crispr/cas pour traiter la dystrophie musculaire de duchenne et la dystrophie musculaire de becker
US11479761B2 (en) 2015-04-16 2022-10-25 Wageningen Universiteit Nuclease-mediated genome editing
US11760985B2 (en) 2015-04-16 2023-09-19 Wageningen Universiteit CRISPR-Cas complex
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
US11180793B2 (en) 2015-04-24 2021-11-23 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US10954513B2 (en) 2015-06-18 2021-03-23 University Of Utah Research Foundation RNA-guided transcriptional regulation and methods of using the same for the treatment of back pain
US11773432B2 (en) 2015-06-18 2023-10-03 The Broad Institute Inc. CRISPR enzymes and systems
US11578312B2 (en) 2015-06-18 2023-02-14 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of CAS9 orthologs and variants for sequence manipulation
JP2021151247A (ja) * 2015-06-18 2021-09-30 ザ・ブロード・インスティテュート・インコーポレイテッド 新規crispr酵素及び系
JP7280312B2 (ja) 2015-06-18 2023-05-23 ザ・ブロード・インスティテュート・インコーポレイテッド 新規crispr酵素及び系
KR20180034402A (ko) * 2015-06-18 2018-04-04 더 브로드 인스티튜트, 인코퍼레이티드 신규한 crispr 효소 및 시스템
US11634755B2 (en) 2015-06-18 2023-04-25 The Broad Institute, Inc. Crispr enzymes and systems
US10876100B2 (en) 2015-06-18 2020-12-29 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US11781172B2 (en) 2015-06-18 2023-10-10 The Broad Institute, Inc. Crispr enzymes and systems
US10494621B2 (en) 2015-06-18 2019-12-03 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
WO2016205688A3 (fr) * 2015-06-18 2017-02-02 Bowles Robert D Régulation de la transcription à guidage arn et procédés d'utilisation de celle-ci pour le traitement des lombalgies
KR102613296B1 (ko) 2015-06-18 2023-12-13 더 브로드 인스티튜트, 인코퍼레이티드 신규한 crispr 효소 및 시스템
EP3800255A3 (fr) * 2015-06-18 2021-06-23 Robert D. Bowles Régulation de la transcription à guidage arn et procédés d'utilisation de celle-ci pour le traitement des lombalgies
US20180155715A1 (en) * 2015-06-18 2018-06-07 Robert D. Bowles Rna-guided transcriptional regulation and methods of using the same for the treatment of back pain
CN105255886A (zh) * 2015-07-09 2016-01-20 青岛市畜牧兽医研究所 一对靶向猪RelA基因的sgRNA
CN105255886B (zh) * 2015-07-09 2018-09-14 青岛市畜牧兽医研究所 一对靶向猪RelA基因的sgRNA
EP3325018A4 (fr) * 2015-07-22 2019-04-24 Duke University Criblage à haut rendement d'une fonction d'élément de régulation à l'aide de technologies d'édition de l'épigénome
US10676735B2 (en) 2015-07-22 2020-06-09 Duke University High-throughput screening of regulatory element function with epigenome editing technologies
WO2017015637A1 (fr) * 2015-07-22 2017-01-26 Duke University Criblage à haut rendement d'une fonction d'élément de régulation à l'aide de technologies d'édition de l'épigénome
EP3325018A1 (fr) * 2015-07-22 2018-05-30 Duke University Criblage à haut rendement d'une fonction d'élément de régulation à l'aide de technologies d'édition de l'épigénome
AU2016311454B2 (en) * 2015-08-25 2023-07-06 Duke University Compositions and methods of improving specificity in genomic engineering using RNA-guided endonucleases
US11427817B2 (en) 2015-08-25 2022-08-30 Duke University Compositions and methods of improving specificity in genomic engineering using RNA-guided endonucleases
AU2016311454C1 (en) * 2015-08-25 2023-10-19 Duke University Compositions and methods of improving specificity in genomic engineering using RNA-guided endonucleases
CN108351350A (zh) * 2015-08-25 2018-07-31 杜克大学 使用rna指导型内切核酸酶改善基因组工程特异性的组合物和方法
CN108351350B (zh) * 2015-08-25 2022-02-18 杜克大学 使用rna指导型内切核酸酶改善基因组工程特异性的组合物和方法
WO2017035416A3 (fr) * 2015-08-25 2017-04-06 Duke University Compositions et procédés d'amélioration de la spécificité dans l'ingénierie génomique à l'aide d'endonucléases guidées par arn
EP4177346A3 (fr) * 2015-08-25 2023-07-26 Duke University Compositions et procédés d'amélioration de la spécificité dans l'ingénierie génomique à l'aide d'endonucléases guidées par arn
US11661583B2 (en) 2015-08-27 2023-05-30 University Of Washington Drug discovery platform for Duchenne cardiomyopathy
WO2017043647A1 (fr) * 2015-09-11 2017-03-16 国立研究開発法人理化学研究所 Procédé de déméthylation de l'adn spécifique à un site de liaison du facteur de transcription
JPWO2017043647A1 (ja) * 2015-09-11 2018-06-28 国立研究開発法人理化学研究所 転写因子結合部位特異的なdna脱メチル化方法
EP3332008A4 (fr) * 2015-09-23 2019-01-16 Université Laval Modification du gène de la dystrophine et ses utilisations
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11421251B2 (en) 2015-10-13 2022-08-23 Duke University Genome engineering with type I CRISPR systems in eukaryotic cells
WO2017066497A3 (fr) * 2015-10-13 2017-06-29 Duke University Ingénierie génomique avec systèmes crispr de type i dans des cellules eucaryotes
US11253576B2 (en) 2015-10-22 2022-02-22 University Of Massachusetts Methods and compositions for treating metabolic imbalance in neurodegenerative disease
US11426469B2 (en) 2015-10-22 2022-08-30 University Of Massachusetts Prostate-targeting adeno-associated virus serotype vectors
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2017075335A1 (fr) 2015-10-28 2017-05-04 Voyager Therapeutics, Inc. Expression régulable au moyen d'un virus adéno-associé (vaa)
EP4279084A1 (fr) * 2015-10-28 2023-11-22 Vertex Pharmaceuticals Inc. Matériaux et méthodes pour traiter la dystrophie musculaire de duchenne
AU2016344609B2 (en) * 2015-10-28 2022-05-12 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of duchenne muscular dystrophy
JP2021126130A (ja) * 2015-10-28 2021-09-02 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated デュシェンヌ型筋ジストロフィーの処置のための材料および方法
US11369692B2 (en) 2015-10-28 2022-06-28 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of Duchenne Muscular Dystrophy
JP2019507579A (ja) * 2015-10-28 2019-03-22 クリスパー セラピューティクス アーゲー デュシェンヌ型筋ジストロフィーの処置のための材料および方法
CN108513546A (zh) * 2015-10-28 2018-09-07 克里斯珀医疗股份公司 用于治疗杜氏肌营养不良症的材料和方法
WO2017072590A1 (fr) * 2015-10-28 2017-05-04 Crispr Therapeutics Ag Matériaux et méthodes pour traiter la dystrophie musculaire de duchenne
CN108779466A (zh) * 2015-11-30 2018-11-09 杜克大学 用于通过基因编辑修正人肌营养不良蛋白基因的治疗靶标和使用方法
JP7108307B2 (ja) 2015-11-30 2022-07-28 デューク ユニバーシティ 遺伝子編集によるヒトジストロフィン遺伝子の修正用の治療標的および使用方法
JP2018534950A (ja) * 2015-11-30 2018-11-29 デューク ユニバーシティ 遺伝子編集によるヒトジストロフィン遺伝子の修正用の治療標的および使用方法
EP3384055A4 (fr) * 2015-11-30 2019-04-17 Duke University Cibles thérapeutiques pour la correction du gène de la dystrophine humaine par l'édition de gènes et procédés d'utilisation
IL259100B2 (en) * 2015-11-30 2023-09-01 Univ Duke Therapeutic targets for human dystrophin gene repair using gene editing and methods for use
CN108779466B (zh) * 2015-11-30 2024-03-29 杜克大学 用于通过基因编辑修正人肌营养不良蛋白基因的治疗靶标和使用方法
IL259100B1 (en) * 2015-11-30 2023-05-01 Univ Duke Therapeutic targets for human dystrophin gene repair through gene editing and methods for use
WO2017123910A1 (fr) * 2016-01-14 2017-07-20 The Brigham And Women's Hospital, Inc. Édition génomique pour traiter un glioblastome
US11826433B2 (en) 2016-02-02 2023-11-28 University Of Massachusetts Method to enhance the efficiency of systemic AAV gene delivery to the central nervous system
JP2019509031A (ja) * 2016-02-11 2019-04-04 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 細胞のゲノムにおける変異ジストロフィン遺伝子を修飾する方法及び組成物
WO2017139505A3 (fr) * 2016-02-11 2017-10-19 The Regents Of The University Of California Procédés et compositions pour modifier un gène mutant de la dystrophine dans le génome d'une cellule
US11666666B2 (en) 2016-02-11 2023-06-06 The Regents Of The University Of California Methods and compositions for modifying a mutant dystrophin gene in a cell's genome
JP6998313B2 (ja) 2016-02-11 2022-02-04 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 細胞のゲノムにおける変異ジストロフィン遺伝子を修飾する方法及び組成物
WO2017139505A2 (fr) 2016-02-11 2017-08-17 The Regents Of The University Of California Procédés et compositions pour modifier un gène mutant de la dystrophine dans le génome d'une cellule
US11060088B2 (en) 2016-02-12 2021-07-13 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11851657B2 (en) 2016-02-12 2023-12-26 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
AU2017235333B2 (en) * 2016-03-14 2023-08-24 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating beta hemoglobinopathies
JP2019508051A (ja) * 2016-03-14 2019-03-28 エディタス・メディシン、インコーポレイテッド β異常ヘモグロビン症を治療するためのCRISPR/CAS関連方法および組成物
CN109153994A (zh) * 2016-03-14 2019-01-04 爱迪塔斯医药公司 用于治疗β-血红蛋白病的CRISPR/CAS相关方法和组合物
KR20180120752A (ko) * 2016-03-14 2018-11-06 에디타스 메디신, 인코포레이티드 베타 이상헤모글로빈증의 치료를 위한 crispr/cas-관련 방법 및 조성물
KR102532663B1 (ko) * 2016-03-14 2023-05-16 에디타스 메디신, 인코포레이티드 베타 이상헤모글로빈증의 치료를 위한 crispr/cas-관련 방법 및 조성물
WO2017160890A1 (fr) * 2016-03-14 2017-09-21 Editas Medicine, Inc. Méthodes et compositions liées à crispr/cas pour traiter les bêta-hémoglobinpathies
EP3219799A1 (fr) 2016-03-17 2017-09-20 IMBA-Institut für Molekulare Biotechnologie GmbH Expression sgrna crispr conditionnelle
WO2017158153A1 (fr) 2016-03-17 2017-09-21 Imba - Institut Für Molekulare Biotechnologie Gmbh Expression conditionnelle de sgrna de crispr
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11207426B2 (en) 2016-04-05 2021-12-28 University Of Massachusetts Compositions and methods for selective inhibition of grainyhead-like protein expression
WO2017180915A3 (fr) * 2016-04-13 2017-11-23 Duke University Répresseurs à base de crispr/cas9 pour inactiver des cibles géniques in vivo et procédés d'utilisation
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11413356B2 (en) 2016-04-15 2022-08-16 University Of Massachusetts Methods and compositions for treating metabolic imbalance
WO2017193029A2 (fr) 2016-05-05 2017-11-09 Duke University Méthodes liées à crispr/cas et compositions destinées à traiter la dystrophie musculaire de duchenne
EP3452498A4 (fr) * 2016-05-05 2019-11-13 Duke University Méthodes liées à crispr/cas et compositions destinées à traiter la dystrophie musculaire de duchenne
JP2019522461A (ja) * 2016-05-05 2019-08-15 デューク ユニバーシティ デュシェンヌ型筋ジストロフィーを治療するためのcrispr/cas関連の方法および組成物
JP7075597B2 (ja) 2016-05-05 2022-05-26 デューク ユニバーシティ デュシェンヌ型筋ジストロフィーを治療するためのcrispr/cas関連の方法および組成物
GB2552861A (en) * 2016-06-02 2018-02-14 Sigma Aldrich Co Llc Using programmable DNA binding proteins to enhance targeted genome modification
US10266851B2 (en) 2016-06-02 2019-04-23 Sigma-Aldrich Co. Llc Using programmable DNA binding proteins to enhance targeted genome modification
GB2552861B (en) * 2016-06-02 2019-05-15 Sigma Aldrich Co Llc Using programmable DNA binding proteins to enhance targeted genome modification
US11882815B2 (en) 2016-06-15 2024-01-30 University Of Massachusetts Recombinant adeno-associated viruses for delivering gene editing molecules to embryonic cells
WO2018009534A1 (fr) * 2016-07-05 2018-01-11 The Johns Hopkins University Compositions et procédés comprenant des améliorations d'arn guides de crispr à l'aide du promoteur h1
CN109844116A (zh) * 2016-07-05 2019-06-04 约翰霍普金斯大学 包括使用h1启动子对crispr指导rna的改进的组合物和方法
US11766488B2 (en) 2016-07-05 2023-09-26 The Johns Hopkins University Compositions and methods comprising improvements of CRISPR guide RNAS using the H1 promoter
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US20200340012A1 (en) * 2016-08-18 2020-10-29 The Regents Of The University Of California Crispr-cas genome engineering via a modular aav delivery system
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11046957B2 (en) 2016-09-22 2021-06-29 University Of Massachusetts AAV treatment of Huntington's disease
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
US11773392B2 (en) 2016-09-22 2023-10-03 University Of Massachusetts AAV treatment of Huntington's disease
US11970710B2 (en) 2016-10-13 2024-04-30 Duke University Genome engineering with Type I CRISPR systems in eukaryotic cells
US11578340B2 (en) 2016-10-13 2023-02-14 University Of Massachusetts AAV capsid designs
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
FR3058160A1 (fr) * 2016-10-28 2018-05-04 Universite Pierre Et Marie Curie - Paris 6 (Upmc) Procede de differenciation de cellules souches pluripotentes en cardiomyocytes
WO2018078301A1 (fr) * 2016-10-28 2018-05-03 Université Pierre Et Marie Curie - Paris 6 (Upmc) Procédé de différenciation de cellules souches pluripotentes en cardiomyocytes
WO2018081978A1 (fr) * 2016-11-03 2018-05-11 深圳华大基因研究院 Méthode et système d'amélioration de l'efficacité de l'édition de gènes
CN110382695A (zh) * 2016-11-28 2019-10-25 得克萨斯州大学系统董事会 通过crispr/cpf1介导的基因编辑来预防肌营养不良
JP2019536782A (ja) * 2016-11-28 2019-12-19 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム CRISPR/Cpf1媒介性遺伝子編集による筋ジストロフィーの予防
WO2018098480A1 (fr) * 2016-11-28 2018-05-31 The Board Of Regents Of The University Of Texas System Prévention de la dystrophie musculaire par édition de gène médiée par crispr/cpf1
US11390652B2 (en) 2016-12-12 2022-07-19 Astellas Pharma Inc. Transcriptional regulatory fusion polypeptide
EP3553090A4 (fr) * 2016-12-12 2020-06-24 Astellas Pharma Inc. Polypeptide fusionné à régulation transcriptionnelle
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
WO2018129296A1 (fr) * 2017-01-05 2018-07-12 The Board Of Regents Of The University Of Texas System Stratégie optimisée pour des modifications par saut d'exon à l'aide de crispr/cas9 avec des séquences de guidage triple
KR102606174B1 (ko) * 2017-01-05 2023-11-27 더 보드 오브 리젠츠 오브 더 유니버시티 오브 텍사스 시스템 트리플 가이드 서열을 지닌 crispr/cas9를 사용한 엑손 스키핑 변형을 위한 최적화된 전략
US20190338311A1 (en) * 2017-01-05 2019-11-07 The Board Of Regents Of The University Of Texas System Optimized strategy for exon skipping modifications using crispr/cas9 with triple guide sequences
KR20190100967A (ko) * 2017-01-05 2019-08-29 더 보드 오브 리젠츠 오브 더 유니버시티 오브 텍사스 시스템 트리플 가이드 서열을 지닌 crispr/cas9를 사용한 엑손 스키핑 변형을 위한 최적화된 전략
JP2020503869A (ja) * 2017-01-05 2020-02-06 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム トリプルガイド配列と共にcrispr/cas9を用いるエクソンスキッピング改変のための最適化戦略
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11591621B2 (en) 2017-02-07 2023-02-28 Modalis Therapeutics Corporation Method of treating diseases associated with elevated KRAS expression using CRISPR-GNDM system
WO2018147343A1 (fr) * 2017-02-07 2018-08-16 Edigene Corporation Méthode de traitement de maladies associées à une expression élevée de kras au moyen d'un système crispr-gndm
WO2018156824A1 (fr) * 2017-02-23 2018-08-30 President And Fellows Of Harvard College Méthodes de modification génétique d'une cellule
US10687520B2 (en) 2017-03-07 2020-06-23 The Board Of Regents Of The University Of Texas System Generation and correction of a humanized mouse model with a deletion of dystrophin exon 44
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
WO2018170184A1 (fr) * 2017-03-14 2018-09-20 Editas Medicine, Inc. Systèmes et méthodes pour le traitement d'hémoglobinopathies
EP3596117A4 (fr) * 2017-03-17 2021-01-13 The Johns Hopkins University Thérapie épigénétique ciblée contre l'élément de régulation distale d'expression du tgfb2
US11771703B2 (en) 2017-03-17 2023-10-03 The Johns Hopkins University Targeted epigenetic therapy against distal regulatory element of TGFβ2 expression
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
CN107012158A (zh) * 2017-03-28 2017-08-04 东南大学 一种端粒酶启动基因表达方法及其应用
CN107012158B (zh) * 2017-03-28 2020-09-11 东南大学 一种端粒酶启动基因表达方法及其应用
CN107058358A (zh) * 2017-04-01 2017-08-18 中国科学院微生物研究所 一种双spacer序列识别切割CRISPR‑Cas9载体构建及其在疣孢菌中的应用
CN107058358B (zh) * 2017-04-01 2020-06-09 中国科学院微生物研究所 一种双spacer序列识别切割CRISPR-Cas9载体构建及其在疣孢菌中的应用
US11345891B2 (en) 2017-04-05 2022-05-31 Asgard Therapeutics Ab Compositions for reprogramming cells into dendritic cells or antigen presenting cells, methods and uses thereof
WO2018185709A1 (fr) 2017-04-05 2018-10-11 Centro De Neurociencias E Biologia Celular Compositions destinées à la reprogrammation de cellules en cellules dendritiques ou en cellules présentatrices d'antigène, procédés et utilisations associés
EP3385373A1 (fr) 2017-04-05 2018-10-10 Centro de Neurociências e Biologia Celular Compositions pour la reprogrammation de cellules en cellules dendritiques ou en cellules présentatrices d'antigènes, leurs procédés et utilisations
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
US11859179B2 (en) 2017-05-09 2024-01-02 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (ALS)
WO2018209158A3 (fr) * 2017-05-10 2019-01-03 Editas Medicine, Inc. Systèmes et procédés de nucléase guidé par crispr/arn
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
US11098297B2 (en) 2017-06-09 2021-08-24 Editas Medicine, Inc. Engineered Cas9 nucleases
WO2019003193A1 (fr) * 2017-06-30 2019-01-03 Novartis Ag Méthodes pour le traitement d'une maladie à l'aide de systèmes d'édition de gènes
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11891603B2 (en) 2017-07-21 2024-02-06 The Governors Of The University Of Alberta Antisense oligonucleotides that bind to exon 51 of human dystrophin pre-mRNA
KR102655989B1 (ko) * 2017-07-21 2024-04-08 더 거버너스 오브 더 유니버시티 오브 알버타 인간 디스트로핀 pre-mRNA의 엑손 51에 결합하는 안티센스 올리고뉴클레오티드
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11739330B2 (en) 2017-09-22 2023-08-29 University Of Massachusetts SOD1 dual expression vectors and uses thereof
US11572574B2 (en) 2017-09-28 2023-02-07 Toolgen Incorporated Artificial genome manipulation for gene expression regulation
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
WO2019131829A1 (fr) 2017-12-28 2019-07-04 国立大学法人京都大学 Composition de modification de gène cible
KR20200104318A (ko) 2017-12-28 2020-09-03 고쿠리츠 다이가쿠 호진 교토 다이가쿠 표적 유전자 개변용 조성물
US11268077B2 (en) 2018-02-05 2022-03-08 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
CN112534054A (zh) * 2018-05-11 2021-03-19 比姆医疗股份有限公司 使用可编程碱基编辑器系统取代病原性氨基酸的方法
WO2020030783A2 (fr) 2018-08-10 2020-02-13 Vib Vzw Moyens et procédés de tolérance à la sécheresse dans des cultures
EP3836974A4 (fr) * 2018-08-18 2022-06-15 President and Fellows of Harvard College Édition génique in situ
WO2020092725A1 (fr) * 2018-11-01 2020-05-07 Montana State University Modulation génique dotée d'un système crispr de type i
WO2020101042A1 (fr) * 2018-11-16 2020-05-22 Astellas Pharma Inc. Méthode de traitement de la dystrophie musculaire par ciblage du gène de l'utrophine
US11473071B2 (en) 2018-11-16 2022-10-18 Astellas Pharma Inc. Method for treating muscular dystrophy by targeting utrophin gene
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
WO2020209800A3 (fr) * 2019-04-11 2020-11-12 Agency For Science, Technology And Research Procédé de modification d'un état de différenciation d'une cellule
CN113785063A (zh) * 2019-04-14 2021-12-10 杜克大学 用于治疗杜氏肌营养不良症的aav载体介导的大规模突变热点缺失
EP3931331A4 (fr) * 2019-04-14 2022-11-30 Duke University Délétion médiée par un vecteur aav d'un point chaud de mutation important pour le traitement de la dystrophie musculaire de duchenne
WO2020237237A1 (fr) * 2019-05-23 2020-11-26 American Molecular Laboratories, Inc. Procédés de détection d'un niveau de h.pylori dans un échantillon fécal
WO2021051054A1 (fr) * 2019-09-13 2021-03-18 The Regents Of The University Of Colorado, A Body Corporate Procédés et compositions pour rajeunissement cellulaire et tissulaire
WO2021058543A1 (fr) * 2019-09-23 2021-04-01 Michalakis Stylianos Procédé de transactivation d'un gène homologue d'un gène d'intérêt et procédé in vitro de diagnostic d'une maladie
WO2021089736A1 (fr) 2019-11-06 2021-05-14 Association Institut De Myologie Polythérapie pour maladies musculaires
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2022008557A2 (fr) 2020-07-08 2022-01-13 UCB Biopharma SRL Modulation de l'expression de cftr
WO2023010135A1 (fr) 2021-07-30 2023-02-02 Tune Therapeutics, Inc. Compositions et procédés pour moduler l'expression de la protéine 2 de liaison méthyle-cpg (mecp2)
WO2023010133A2 (fr) 2021-07-30 2023-02-02 Tune Therapeutics, Inc. Compositions et procédés de modulation de l'expression de la frataxine
CN113637658A (zh) * 2021-08-19 2021-11-12 中国农业科学院棉花研究所 基于dCas9-oToV的基因转录系统及其应用
WO2023137471A1 (fr) 2022-01-14 2023-07-20 Tune Therapeutics, Inc. Compositions, systèmes et procédés de programmation de phénotypes de lymphocytes t par activation génique ciblée
WO2023137472A2 (fr) 2022-01-14 2023-07-20 Tune Therapeutics, Inc. Compositions, systèmes et procédés de programmation de phénotypes de lymphocytes t par répression génique ciblée
EP4215614A1 (fr) 2022-01-24 2023-07-26 Dynacure Thérapie de combinaison pour maladies liées à la dystrophine
WO2023250511A2 (fr) 2022-06-24 2023-12-28 Tune Therapeutics, Inc. Compositions, systèmes et procédés de réduction de lipoprotéine de faible densité par répression génique ciblée
WO2023247789A1 (fr) 2022-06-24 2023-12-28 European Molecular Biology Laboratory Outil modulaire basé sur une crispr pour l'introduction spécifique de modifications épigénétiques sur des loci cibles
WO2024015881A2 (fr) 2022-07-12 2024-01-18 Tune Therapeutics, Inc. Compositions, systèmes et procédés d'activation transcriptionnelle ciblée
WO2024040254A2 (fr) 2022-08-19 2024-02-22 Tune Therapeutics, Inc. Compositions, systèmes et méthodes de régulation du virus de l'hépatite b par répression génique ciblée
WO2024064642A2 (fr) 2022-09-19 2024-03-28 Tune Therapeutics, Inc. Compositions, systèmes et méthodes de modulation de fonction de lymphocyte t

Also Published As

Publication number Publication date
US20160201089A1 (en) 2016-07-14
WO2014197748A3 (fr) 2015-02-19
KR20210072831A (ko) 2021-06-17
CN105658805B (zh) 2021-12-31
EP3539573A1 (fr) 2019-09-18
EP3539573B1 (fr) 2024-02-14
CN114230675A (zh) 2022-03-25
US20180320197A1 (en) 2018-11-08
AU2014274840A2 (en) 2016-01-21
KR102307280B1 (ko) 2021-10-01
AU2022250450A1 (en) 2023-01-05
US20210002665A1 (en) 2021-01-07
AU2014274840B2 (en) 2020-03-12
AU2020203924A1 (en) 2020-07-02
JP2023164823A (ja) 2023-11-14
KR20160023765A (ko) 2016-03-03
US20210032654A1 (en) 2021-02-04
KR20230107387A (ko) 2023-07-14
AU2014274840A1 (en) 2016-01-21
US10704060B2 (en) 2020-07-07
US10745714B2 (en) 2020-08-18
EP3417880A1 (fr) 2018-12-26
JP7313055B2 (ja) 2023-07-24
JP7085716B2 (ja) 2022-06-17
AU2022250572A1 (en) 2023-01-05
EP3004370A4 (fr) 2017-01-11
EP3004370A2 (fr) 2016-04-13
CN105658805A (zh) 2016-06-08
JP2016521555A (ja) 2016-07-25
CA2914519A1 (fr) 2014-12-11
JP2022101562A (ja) 2022-07-06
KR102551324B1 (ko) 2023-07-05
JP2020058349A (ja) 2020-04-16
KR20230136697A (ko) 2023-09-26

Similar Documents

Publication Publication Date Title
US10745714B2 (en) RNA-guided gene editing and gene regulation
US20210040460A1 (en) Genetic correction of mutated genes
EP3487523B1 (fr) Applications thérapeutiques de l'édition du génome fondée sur cpf1
EP3452498B1 (fr) Composiitons liées à crispr/cas destinées à traiter la dystrophie musculaire de duchenne
JP2022101562A5 (fr)
JP2019530467A (ja) 安全性向上のための自己制限Cas9回路網(SLiCES)プラスミドおよびそのレンチウイルス系
US9738879B2 (en) Genetic correction of mutated genes
JP2023522788A (ja) 標的化されたゲノム組込みによってデュシェンヌ型筋ジストロフィーを矯正するためのcrispr/cas9療法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14806852

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2914519

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016518017

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20167000166

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2014806852

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2014274840

Country of ref document: AU

Date of ref document: 20140605

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14806852

Country of ref document: EP

Kind code of ref document: A2